EP1888649A2 - Antigen-bindungsmoleküle mit modifizierten fc-regionen und veränderte bindung an fc-rezeptoren - Google Patents

Antigen-bindungsmoleküle mit modifizierten fc-regionen und veränderte bindung an fc-rezeptoren

Info

Publication number
EP1888649A2
EP1888649A2 EP06831545A EP06831545A EP1888649A2 EP 1888649 A2 EP1888649 A2 EP 1888649A2 EP 06831545 A EP06831545 A EP 06831545A EP 06831545 A EP06831545 A EP 06831545A EP 1888649 A2 EP1888649 A2 EP 1888649A2
Authority
EP
European Patent Office
Prior art keywords
antigen binding
binding molecule
glycoengineered
molecule according
antibody
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP06831545A
Other languages
English (en)
French (fr)
Inventor
Peter Sondermann
Claudia Ferrara Koller
Peter Brunker
Fiona Stuart
Pablo Umana
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Roche Glycart AG
Original Assignee
Roche Glycart AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Roche Glycart AG filed Critical Roche Glycart AG
Publication of EP1888649A2 publication Critical patent/EP1888649A2/de
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/40Immunoglobulins specific features characterized by post-translational modification
    • C07K2317/41Glycosylation, sialylation, or fucosylation

Definitions

  • the present invention is directed to antigen binding molecules, including antibodies, comprising a Fc region having one or more amino acid modifications, wherein the antigen binding molecule exhibits altered binding to one or more Fc receptors as a result of the modification(s).
  • the invention is further directed to polynucleotides and vectors encoding such antigen binding molecules, to host cells comprising same, to methods for making the antigen binding molecules of the invention, and to their use in the treatment of various diseases and disorders, e.g., cancers.
  • Antibodies provide a link between the humoral and the cellular immune system with IgG being the most abundant serum immunoglobulin. While the Fab regions of the antibody recognize antigens, the Fc portion binds to Fc ⁇ receptors (Fc ⁇ Rs) that are differentially expressed by all immune competent cells. Upon receptor crosslinking by a multivalent antigen/antibody complex, degranulation, cytolysis or phagocytosis of the target cell and transcriptional-activation of cytokine-encoding genes are triggered (Deo, Y.M. et al, Immunol. Today 18(3):127-135 (1997)).
  • effector functions mediated by the antibody Fc region can be divided into two categories: (1) effector functions that operate after the binding of antibody to an antigen (these functions involve, for example, the participation of the complement cascade or Fc receptor (FcR)-bearing cells); and (2) effector functions that operate independently of antigen binding (these functions confer, for example, persistence in the circulation and the ability to be transferred across cellular barriers by transcytosis).
  • effector functions that operate after the binding of antibody to an antigen (these functions involve, for example, the participation of the complement cascade or Fc receptor (FcR)-bearing cells); and (2) effector functions that operate independently of antigen binding (these functions confer, for example, persistence in the circulation and the ability to be transferred across cellular barriers by transcytosis).
  • FcR complement cascade or Fc receptor
  • FcR Fc receptor
  • IgG gamma receptors
  • IgE epsilon receptors
  • IgA alpha receptors
  • IgM mi receptors
  • binding of antibody to Fc receptors on cell surfaces triggers a number of important and diverse biological responses including engulfhient and destruction of antibody-coated particles, clearance of immune complexes, lysis of antibody-coated target cells by killer cells (known as antibody-dependent cell-mediated cytotoxicity, or ADCC), release of inflammatory mediators, placental transfer and control of immunoglobulin production.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • FcRs are defined by their specificity for immunoglobulin isotypes; Fc receptors for IgG antibodies are referred to as Fc ⁇ R, for IgE as Fc ⁇ R, for IgA as Fc ⁇ R and so on. Three subclasses of human Fc ⁇ R have been identified: Fc ⁇ RI (CD64), Fc ⁇ RII (CD32) and Fc ⁇ RIII (CD16).
  • the three genes encoding the Fc ⁇ RI subclass (Fc ⁇ RIA, Fc ⁇ RIB and Fc ⁇ RIC) are clustered in region Iq21.1 of the long arm of chromosome 1; the genes encoding Fc ⁇ RII isoforms (Fc ⁇ RIIA, Fc ⁇ RIIB and Fc ⁇ RIIC) and the two genes encoding Fc ⁇ RIII (Fc ⁇ RIIIA and Fc ⁇ RIIIB) are all clustered in region Iq22.
  • These different FcR subtypes are expressed on different cell types ⁇ see, e.g., Ravetch, J.V. and Kinet, J.P.
  • Fc ⁇ RIIIB is found only on neutrophils
  • Fc ⁇ RIIIA is found on macrophages, monocytes, natural killer (NK) cells, and a subpopulation of T-cells.
  • Fc ⁇ RIIIA is the only FcR present on NK cells, one of the cell types implicated in ADCC.
  • Fc ⁇ RI, Fc ⁇ RII and Fc ⁇ RIII are immunoglobulin superfamily (IgSF) receptors;
  • Fc ⁇ RI has three IgSF domains in its extracellular domain, while Fc ⁇ RII and Fc ⁇ RIH have only two IgSF domains in their extracellular domains.
  • FcRn neonatal Fc receptor
  • MHC major histocompatibility complex
  • the N- glycosylation pattern of mAbs has been manipulated by engineering the glycosylation pathway of a production cell line using enzyme activities that lead to naturally occurring carbohydrates.
  • the resulting glycoengineered (GE) antibodies feature high proportions of bisected, non-fucosylated oligosaccharides, improved affinity for Fc ⁇ RIIIa and enhanced ADCC (Umana, P. et al, Nat. Biotech. 17 (2) :17 '6-180 (1999), Ferrara, C. et al, submitted). Similar results are found using a production cell line which is unable to add fucose residues to N-linked oligosaccharides (Sarmay, G. et ah, MoI. Immunol. 29(5):633-639 (1992).
  • the invention is directed to a glycoengineered antigen-binding molecule comprising a Fc region, wherein said Fc region has an altered oligosaccharide structure as a result of said glycoengineering and has at least one amino acid modification, and wherein said antigen binding molecule exhibits increased binding to a human Fc ⁇ RIII receptor compared to the antigen binding molecule that lacks said modification.
  • the glycoengineered antigen binding molecule does not exhibit increased binding to a human Fc ⁇ RII receptor, such as the Fc ⁇ RIIa receptor or the Fc ⁇ RIIb receptor.
  • the Fc ⁇ RIII receptor is glycosylated such that it comprises N-linked oligosaccharides at Asnl62.
  • the Fc ⁇ RIII receptor is Fc ⁇ RIIIa.
  • the Fc ⁇ RIII receptor is Fc ⁇ RIIIb.
  • the Fc ⁇ RIIIa receptor has a valine residue at position 158. hi other embodiments, the Fc ⁇ RIIIa receptor has a phenylalanine residue at position 158.
  • the glycoengineered antigen binding molecule of the present invention contains a modification that does not substantially increase binding to a nonglycosylated Fc ⁇ RIII receptor compared to the antigen binding molecule lacking said modification.
  • the glycoengineered antigen binding molecule of the present invention comprises a substitution at one or more of amino acids 239, 241, 243, 260, 262, 263, 264, 265, 268, 290, 292, 293, 294, 295, 296, 297, 298, 299, 300, 301, 302, or 303.
  • the glycoengineered antigen binding molecule comprises two or more of the substitutions listed in Tables 2 and 4.
  • the glycoengineered antigen binding molecule comprises the two or more substitutions listed in Table 5.
  • the present invention is further directed to a glycoengineered antigen binding molecule comprising one or more substitutions that replace the naturally occurring amino acid residue with an amino acid residue that interacts with the carbohydrate attached to Asnl62 of the Fc ⁇ RIII receptor.
  • the amino acid residue that interacts with the carbohydrate attached to Asnl62 of the Fc ⁇ RIII receptor is selected from the- group consisting of: Trp, His, Tyr, GIu, Arg, Asp, Phe, Asn, and GIn.
  • the glycoengineered antigen binding molecule comprises a substitution selected from the group consisting of: Ser239Asp, Ser239Glu, Ser239Trp, Phe243His, Phe243Glu, Thr260His, His268Asp, His268Glu.
  • the glycoengineered antigen binding molecule according to the present invention may contain one or more substitutions listed in Tables 2 or 4.
  • the glycoengineered antigen binding molecule of the present invention binds to the Fc ⁇ RIII receptor with at least 10 % increased affinity, at least 20% increased affinity, at least 30% increased affinity, at least 40% increased affinity, at least 50% increased affinity, at least 60% increased affinity, at least 70% increased affinity, at least 80% increased affinity, at least 90%, increased affinity, or at least 100% increased affinity compared to the same antigen binding molecule lacking said modification.
  • the glycoengineered antigen binding molecule of the present invention preferably comprises a human IgG Fc region.
  • the antigen binding molecule is an antibody or an antibody fragment comprising an Fc region.
  • the antibody or antibody fragment is chimeric or humanized.
  • the glycoengineered antigen binding molecule according to the invention exhibits increased effector function.
  • the increased effector function is increased antibody-dependent cellular cytotoxicity or increased complement dependent cytotoxicity.
  • the altered oligosaccharide structure in the glycoengineered antigen binding molecules of the present invention preferably comprises a decreased number of fucose residues as compared to the nonglycoengineered antigen binding molecule.
  • at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80% or more of the oligosaccharides in the Fc region are nonfucosylated.
  • the altered oligosaccharide structure in the glycoengineered antigen binding molecules of the present invention may also comprise an increased number of bisected oligosaccharides as compared to the nonglycoengineered antigen binding molecule.
  • the bisected oligosaccharide may be of the hybrid type or the complex type.
  • the present invention also encompasses a glycoengineered antigen binding molecule, wherein said altered oligosaccharide structure comprises an increase in the ratio of GIcNAc residues to , fucose residues as compared to the nonglycoengineered antigen binding molecule.
  • the glycoengineered antigen binding molecules of the present invention selectively bind an antigen selected from the group consisting of: the human CD20 antigen, the human EGFR antigen, the human MCSP antigen, the human MUC-I antigen, the human CEA antigen, the human HER2 antigen, and the human TAG- 72 antigen.
  • the present invention is also directed to a glycoengineered antigen binding molecule comprising a Fc region, wherein said Fc region has an altered oligosaccharide structure as a result of said glycoengineering and has at least one amino acid modification, and wherein said antigen binding molecule exhibits increased specificity to a human Fc ⁇ RIII receptor compared to the antigen binding molecule that lacks said modification.
  • the glycoengineered antigen binding molecule of the present invention does not exhibit increased specificity to a human Fc ⁇ RII receptor, such as the human Fc ⁇ RIIa receptor or the human Fc ⁇ RIIb receptor.
  • the Fc ⁇ RIII receptor is preferably glycosylated, (i.e., it comprises N-linked oligosaccharides at Asnl62).
  • the Fc ⁇ RIII receptor is Fc ⁇ RIIIa.
  • the Fc ⁇ RIII receptor is Fc ⁇ RIIIb.
  • the Fc ⁇ RIIIa receptor has a valine residue at position 158.
  • the Fc ⁇ RIIIa receptor has a phenylalanine residue at position 158.
  • the amino acid modification of an antigen binding molecule does not substantially increase specificity for a nonglycosylated Fc ⁇ RIII receptor compared to the antigen binding molecule lacking the modification.
  • the modification comprises an amino acid substitution at one or more of amino acid positions 239, 241, 243, 260, 262, 263, 264, 265, 268, 290, 292, 293, 294, 295, 296, 297, 298, 299, 300, 301, 302, or 303.
  • the substitution replaces the naturally occurring amino acid residue with an amino acid residue that interacts with the carbohydrate attached to Asnl62 of the Fc ⁇ RIII receptor.
  • the amino acid residue that interacts with the carbohydrate attached to Asnl62 of the Fc ⁇ RIII receptor is selected from the group consisting of: Trp, His, Tyr, GIu, Arg, Asp, Phe, Asn, and GIn.
  • substitution is selected from the group consisting of:
  • glycoengineered antigen binding molecule may also contain one or more of the substitutions listed in Tables 2 or 5.
  • the invention encompasses a glycoengineered antigen binding molecule wherein said antigen binding molecule binds to a Fc ⁇ RIII receptor with at least 10% increased specificity, at least 20% increased specificity, at least 30% increased specificity, at least 40% increased specificity, at least 50% increased specificity, at least 60% increased specificity, at least 70% increased specificity, at least 80% increased specificity, at least 90% increased specificity, or at least 100% increased specificity or more compared to the antigen binding molecule lacking said modification.
  • the glycoengineered antigen binding molecule of the invention exhibiting increased specificity contains a human IgG Fc region.
  • the antigen binding molecule is an antibody or an antibody fragment comprising an Fc region.
  • the antibody or antibody fragment is chimeric or humanized.
  • the glycoengineered antigen binding molecule according to the invention preferably exhibits increased effector function, e.g., increased antibody-dependent cellular cytotoxicity or increased complement dependent cytotoxicity.
  • the altered oligosaccharide structure may comprise a decreased number of fucose residues as compared to the nonglycoengineered antigen binding molecule.
  • the invention encompasses a glycoengineered antigen binding molecule, wherein at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90% or more of the oligosaccharides in the Fc region are nonfucosylated.
  • the altered oligosaccharide structure may comprise an increased number of bisected oligosaccharides as compared to the nonglycoengineered antigen binding molecule.
  • the bisected oligosaccharides may be of hybrid type or the complex type, m one embodiment, the altered oligosaccharide structure comprises an increase in the ratio of GIcNAc residues to fucose residues as compared to the nonglycoengineered antigen binding molecule.
  • the glycoengineered antigen binding molecules according to the invention selectively bind an antigen selected from the group consisting of: the human CD20 antigen, the human EGFR antigen, the human MCSP antigen, the human MUC-I antigen, the human CEA antigen, the human HER2 antigen; and the human TAG-72 antigen.
  • the present invention is also directed to a polynucleotide encoding a polypeptide comprising an antibody Fc region or a fragment of an antibody Fc region, wherein said Fc region or fragment thereof has at least one amino acid modification, and wherein said polypeptide exhibits increased binding to a human Fc ⁇ RIII receptor compared to the same polypeptide that lacks said modification.
  • the present invention is also directed to polypeptides encoded by such polynucleotides.
  • the polypeptide may be an antibody heavy chain.
  • the polypeptide may also be fusion protein.
  • the present invention is further directed to vectors and host cells comprising the polynucleotides of the invention.
  • the present invention is also directed to a method for producing a glycoengineered antigen binding molecule comprising a Fc region, wherein said Fc region has an altered oligosaccharide structure as a result of said glycoengineering and has at least one amino acid modification, and wherein said antigen binding molecule exhibits increased binding to a human Fc ⁇ RIII receptor compared to the antigen binding molecule that lacks said modification, said method comprising:
  • the invention is also directed to a method for producing a glycoengineered antigen binding molecule comprising a Fc region, wherein said Fc region has an altered oligosaccharide structure as a result of said glycoengineering and has at least one amino acid modification, and wherein said antigen binding molecule exhibits increased selectivity to a human Fc ⁇ RIII receptor compared to the antigen binding molecule that lacks said modification, said method comprising: (i) culturing the host cell of the invention under conditions permitting the expression of said polynucleotide; and (ii) recovering said glycoengineered antigen binding molecule from the culture medium.
  • Glyco-1 refers to a glycoengineered antibody variant generated from overexpression of GnT-III alone.
  • Glyco-2 refers to a glycoengineered antibody variant generated by co-expression of GnT-III and recombinant ManII.
  • Fig. 2(a-b) Binding of the shFc ⁇ RIIIa[Val-158] or shFc ⁇ RIUa[Phe-158] to immobilized IgGl glycovariants.
  • the association phase is represented by a solid bar above the curves, (a) Overlay of sensograms of the binding events for shFc ⁇ RHIa[Val- 158] and shFc ⁇ RIIIa[Phe-158], respectively. To compare the binding event of GE antibodies within a similar response range, the sensograms obtained at concentrations of 800 nM or 6.4 ⁇ M for the native antibody were overlaid.
  • Fig. 4(a-b) The proposed interaction of the glycosylated Fc ⁇ RIII with the Fc- fragment of IgG.
  • the two chains of the Fc fragment and the unglycosylated Fc ⁇ RIII are depicted as surface with Asnl62 and the fucose residue indicated.
  • the glycans attached to the Fc are shown as ball and sticks.
  • the fucose residue linked to the carbohydrate of the Fc fragment chain is responsible for the sterical hindrance of the proposed interaction with the Fc ⁇ RIII carbohydrate, (b) Model of interaction between a glycosylated Fc ⁇ RIII and the (non-fucosylated) Fc fragment of GE-IgG.
  • the fucose residue is not present within GE-IgG, the carbohydrates attached at Asnl62 of the receptor can thoroughly interact with the GE-IgG.
  • the figure was created using the program PYMOL (www.delanoscientific.com).
  • ABBREVIATIONS Ig, Immunoglobulin; ADCC 5 Antibody-dependent cellular cytotoxicity; CDC, Complement-dependant cytotoxicity; PBMC, Peripheral blood mononuclear cells; GE, Glyco-engineered; GIcNAc, N-Acetylglucosamine; Man, mannose; Gal, galactose; Fuc, fucose; NeuAc, N-acetylneuraminic acid; GnT-III, N- acetylglucosaminyltransferase III; Ic 0n , association rate constant; k off , dissociation rate constant.
  • antibody is intended to include whole antibody molecules, including monoclonal, polyclonal and multispecific (e.g., bispecific) antibodies, as well as antibody fragments having the Fc region and retaining binding specificity and at least one effector function, e.g., ADCC, and fusion proteins that include a region functionally equivalent to the Fc region of an immunoglobulin and that retain binding specificity and at least one effector function. Also encompassed are chimeric and humanized antibodies, as well as camelized and primatized antibodies.
  • Fc region is intended to refer to a C-terminal region of a human IgG heavy chain. Although the boundaries of the Fc region of an IgG heavy chain might vary slightly, the human IgG heavy chain Fc region is usually defined to stretch from the amino acid residue at position Cys226 to the carboxyl-terminus.
  • region equivalent to the Fc region of an immunoglobulin is intended to include naturally occurring allelic variants of the Fc region of an immunoglobulin as well as variants having alterations which produce substitutions, additions, or deletions but which do not decrease substantially the ability of the immunoglobulin to mediate effector functions (such as antibody dependent cellular cytotoxicity).
  • one or more amino acids can be deleted from the N-terminus or C-terminus of the Fc region of an immunoglobulin without substantial loss of biological function.
  • variants can be selected according to general rules known in the art so as to have minimal effect on activity. (See, e.g., Bowie, J. U. et al., Science 247:1306-10 (1990)).
  • the term antigen binding molecule or ABM refers in its broadest sense to a molecule that specifically binds an antigenic determinant.
  • the ABM is an antibody; however, single chain antibodies, single chain Fv molecules, Fab fragments, diabodies, triabodies, tetrabodies, and the like are also contemplated by the present invention.
  • fusion and chimeric when used in reference to polypeptides such as ABMs refer to polypeptides comprising amino acid sequences derived from two or more heterologous polypeptides, such as portions of antibodies from different species.
  • the non-antigen binding components may be derived from a wide variety of species, including primates such as chimpanzees and humans.
  • the constant region of the chimeric ABM is most preferably substantially identical to the constant region of a natural human antibody; the variable region of the chimeric antibody is most preferably substantially identical to that of a recombinant antibody having the amino acid sequence of the murine variable region.
  • Humanized antibodies are a particularly preferred form of fusion or chimeric antibody.
  • a polypeptide having, for example, GnT-III activity refers to a polypeptide that is able to catalyze the addition of a N-acetylglucosamine (GIcNAc) residue in ⁇ -1-4 linkage to the ⁇ -linked mannoside of the trimannosyl core of'Nrlinked oligosaccharides.
  • GIcNAc N-acetylglucosamine
  • ⁇ (l,4)-N-acetylglucosaminyltransferase III also known as ⁇ -l,4-mannosyl-glycoprotein 4- ⁇ -N-acetylglucosaminyl-transferase (EC 2.4.1.144)
  • NC-IUBMB Nomenclature Committee of the International Union of Biochemistry and Molecular Biology
  • the candidate polypeptide will exhibit greater activity or not more than about 25 fold less and, preferably, not more than about tenfold less activity, and most preferably, not more than about three fold less activity relative to the GnT-III.
  • variant refers to a polypeptide differing from a specifically recited polypeptide of the invention by amino acid insertions, deletions, and substitutions, created using, e g., recombinant DNA techniques.
  • variants of the ABMs of the present invention include chimeric, primatized, or humanized antigen binding molecules wherein one or several of the amino acid residues are modified by substitution, addition and/or deletion in such manner that does not substantially affect antigen binding affinity or antibody effector function.
  • Guidance in determining which amino acid residues may be replaced, added, or deleted without abolishing activities of interest may be found by comparing the sequence of the particular polypeptide with that of homologous peptides and minimizing the number of amino acid sequence changes made in regions of high homology (conserved regions) or by replacing amino acids with consensus sequences.
  • recombinant variants encoding these same or similar polypeptides may be synthesized or selected by making use of the "redundancy" in the genetic code.
  • Various codon substitutions such as the silent changes which produce various restriction sites, may be introduced to optimize cloning into a plasmid or viral vector or expression in a particular prokaryotic or eukaryotic system.
  • Mutations in the polynucleotide sequence may be reflected in the polypeptide or domains of other peptides added to the polypeptide to modify the properties of any part of the polypeptide, to change characteristics such as ligand-binding affinities, interchain affinities, or degradation/turnover rate.
  • amino acid substitutions are the result of replacing one amino acid with another amino acid having similar structural and/or chemical properties, i.e., conservative amino acid replacements.
  • conservative amino acid replacements may be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues involved.
  • nonpolar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan, and methionine; polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine, and glutamine; positively charged (basic) amino acids include arginine, lysine, and histidine; and negatively charged (acidic) amino acids include aspartic acid and glutamic acid.
  • “Insertions” or “deletions” are preferably in the range of about 1 to about 20 amino acids, more preferably about 1 to about 10 amino acids. The variation allowed may be experimentally determined by systematically making insertions, deletions, or substitutions of amino acids in a polypeptide molecule using recombinant DNA techniques and assaying the resulting recombinant variants for activity.
  • humanized is used to refer to an antigen binding molecule (ABM) derived from a non-human antigen binding molecule, for example, a murine antibody, that retains or substantially retains the antigen binding properties of the parent molecule but which is less immunogenic in humans.
  • ABSM antigen binding molecule
  • This may be achieved by various methods including (a) grafting only the non-human complementarity determining regions (CDRs) onto human framework and constant regions with or without retention of critical framework residues (e.g., those that are important for retaining good antigen binding affinity or antibody functions), or (b) transplanting the entire non-human variable domains, but "cloaking" them with a human-like section by replacement of surface residues.
  • CDRs complementarity determining regions
  • CDRl There are generally three CDRs (CDRl, CDR2, and CDR3) in each of the heavy and light chain variable domains of an antibody, which are flanked by four framework subregions (i.e., FRl, FR2, FR3, and FR4): FRl -CDRl -FR2-CDR2-FR3-CDR3-FR4.
  • primatized is used to refer to an antigen binding molecule derived from a non-primate antigen binding molecule, for example, a murine antibody, that retains or substantially retains the antigen binding properties of the parent molecule but which is less immunogenic in primates.
  • CDR complementarity determining region
  • Kabat et al. also defined a numbering system for variable domain sequences that is applicable to any antibody.
  • Kabat numbering refers to the numbering system set forth in Kabat et ah, U.S. Dept. of Health and Human Services, "Sequence of Proteins of Immunological Interest” (1983) (incorporated herein by reference in its entirety).
  • the sequences of any sequence listing i.e., SEQ ID NO:1 to SEQ ID NO:2 are not numbered according to the Kabat numbering system. However, as stated above, it is well within the ordinary skill of one in the art to determine the Kabat numbering scheme of any variable region sequence in the Sequence Listing based on the numbering of the sequences as presented therein.
  • nucleic acid or polynucleotide having a nucleotide sequence at least, for example, 95% identical, or having 95% identity, to a reference nucleotide sequence of the present invention it is intended that the nucleotide sequence of the polynucleotide is identical to the reference sequence except that the polynucleotide sequence may include up to five point mutations per each 100 nucleotides of the reference nucleotide sequence.
  • nucleotide sequence at least 95% identical to a reference nucleotide sequence up to 5% of the nucleotides in the reference sequence may be deleted or substituted with another nucleotide, or a number of nucleotides up to 5% of the total nucleotides in the reference sequence may be inserted into the reference sequence.
  • nucleic acid molecule or polypeptide is at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to a nucleotide sequence or polypeptide sequence of the present invention can be determined conventionally using known computer programs.
  • a preferred method for determining the best overall match between a query sequence (a sequence of the present invention) and a subject sequence, also referred to as a global sequence alignment, can be determined using the FASTDB computer program based on the algorithm of Brutlag et ah, Comp. App. Biosci. (5:237-245 (1990). hi a sequence alignment the query and subject sequences are both DNA sequences.
  • RNA sequence can be compared by converting U's to T's.
  • the result of said global sequence alignment is in percent identity.
  • the percent identity is corrected by calculating the number of bases of the query sequence that are 5' and 3' of the subject sequence, which are not matched/aligned, as a percent of the total bases of the query sequence. Whether a nucleotide is matched/aligned is determined by results of the FASTDB sequence alignment.
  • This percentage is then subtracted from the percent identity, calculated by the above FASTDB program using the specified parameters, to arrive at a final percent identity score.
  • This corrected score is what is used for the purposes of the present invention. Only bases outside the 5' and 3' bases of the subject sequence, as displayed by the FASTDB alignment, which are not matched/aligned with the query sequence, are calculated for the purposes of manually adjusting the percent identity score.
  • a 90 base subject sequence is aligned to a 100 base query sequence to determine percent identity.
  • the deletions occur at the 5' end of the subject sequence and therefore, the FASTDB alignment does not show a matched/alignment of the first 10 bases at 5' end.
  • the 10 unpaired bases represent 10% of the sequence (number of bases at the 5' and 3' ends not matched/total number of bases in the query sequence) so 10% is subtracted from the percent identity score calculated by the FASTDB program. If the remaining 90 bases were perfectly matched the final percent identity would be 90%.
  • a 90 base subject sequence is compared with a 100 base query sequence.
  • deletions are internal deletions so that there are no bases- on the 5 ' or 3' end of the subject sequence which are not matched/aligned with the query.
  • percent identity calculated by FASTDB is not manually corrected.
  • bases on the 5' and 3' end of the subject sequence which are not matched/aligned with the query sequence are manually corrected for. No other manual corrections are to made for the purposes of the present invention.
  • polypeptide having an amino acid sequence at least, for example, 95%
  • amino acid sequence of the subject polypeptide is identical to the query sequence except that the subject polypeptide sequence may include up to five amino acid alterations per each 100 amino acids of the query amino acid sequence.
  • the subject polypeptide sequence may include up to five amino acid alterations per each 100 amino acids of the query amino acid sequence.
  • up to 5% of the amino acid residues in the subject sequence may be inserted, deleted, or substituted with another amino acid.
  • These alterations of the reference sequence may occur at the amino or carboxy terminal positions of the reference amino acid sequence or anywhere between those terminal positions, interspersed either individually among residues in the reference sequence or in one or more contiguous groups within the reference sequence.
  • a preferred method for determining the best overall match between a query sequence (a sequence of the present invention) and a subject sequence can be determined using the FASTDB computer program based on the algorithm of Brutlag et ah, Comp. App. Biosci. (5:237-245 (1990).
  • a sequence alignment the query and subject sequences are either both nucleotide sequences or both amino acid sequences. The result of said global sequence alignment is in percent identity.
  • the percent identity is corrected by calculating the number of residues of the query sequence that are N- and C-terminal of the subject sequence, which are not matched/aligned with a corresponding subject residue, as a percent of the total bases of the query sequence. Whether a residue is matched/aligned is determined by results of the FASTDB sequence alignment.
  • This percentage is then subtracted from the percent identity, calculated by the above FASTDB program using the specified parameters, to arrive at a final percent identity score.
  • This final percent identity score is what is used for the purposes of the present invention. Only residues to the N- and C-termini of the subject sequence, which are not matched/aligned with the query sequence, are considered for the purposes of manually adjusting the percent identity score. That is, only query residue positions outside the farthest N- and C-terminal residues of the subject sequence.
  • a 90 amino acid residue subject sequence is aligned with a 100 residue query sequence to determine percent identity.
  • the deletion occurs at the N- terminus of the subject sequence and therefore, the FASTDB alignment does not show a matching/alignment of the first 10 residues at the N-terminus.
  • the 10 unpaired residues represent 10% of the sequence (number of residues at the N- and C-termini not matched/total number of residues in the query sequence) so 10% is subtracted from the percent identity score calculated by the FASTDB program. If the remaining 90 residues were perfectly matched the final percent identity would be 90%.
  • a 90 residue subject sequence is compared with a 100 residue query sequence.
  • deletions are internal deletions so there are no residues at the N- or C-termini of the subject sequence which are not matched/aligned with the query.
  • percent identity calculated by FASTDB is not manually corrected.
  • residue positions outside the N— and C-terminal ends of the subject sequence, as displayed in the FASTDB alignment, which are not matched/aligned with the query sequence are manually corrected for. No other manual corrections are to be made for the purposes of the present invention.
  • a nucleic acid that hybridizes under stringent conditions to a nucleic acid sequence of the invention refers to a polynucleotide that hybridizes in an overnight incubation at 42° C in a solution comprising 50% formamide, 5x SSC (750 niM NaCl, 75 mM sodium citrate), 50 mM sodium phosphate (pH 7.6), 5x Denhardt's solution, 10% dextran sulfate, and 20 ⁇ g/ml denatured, sheared salmon sperm DNA, followed by washing the filters in O.lx SSC at about 65 0 C.
  • 5x SSC 750 niM NaCl, 75 mM sodium citrate
  • 50 mM sodium phosphate pH 7.6
  • 5x Denhardt's solution 10% dextran sulfate
  • 20 ⁇ g/ml denatured, sheared salmon sperm DNA followed by washing the filters in O.lx SSC at about 65 0 C.
  • Golgi localization domain refers to the amino acid sequence of a Golgi resident polypeptide which is responsible for, anchoring the polypeptide in location within the Golgi complex.
  • localization domains comprise amino terminal "tails" of an enzyme.
  • effector function refers to those biological activities attributable to the Fc region (a native sequence Fc region or amino acid sequence variant Fc region) of an antibody.
  • antibody effector functions include, but are not limited to, Fc receptor binding affinity, antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), cytokine secretion, immune-complex- mediated antigen uptake by antigen-presenting cells, down-regulation of cell surface receptors, etc.
  • the terms engineer, engineered, engineering, glycoengineer, glycoengineered, glycoengineering, and glycosylation engineering are considered to include any manipulation of the glycosylation pattern of a naturally occurring or recombinant polypeptide, such as an antigen binding molecule (ABM), or fragment thereof.
  • Glycosylation engineering includes metabolic engineering of the glycosylation machinery of a cell, including genetic manipulations of the oligosaccharide synthesis pathways to achieve altered glycosylation of glycoproteins expressed in cells.
  • glycosylation engineering includes the effects of mutations and cell environment on glycosylation.
  • the glycosylation engineering is an alteration in glycosyltransferase activity.
  • the engineering results in altered glucosaminyltransferase activity and/or fucosyltransferase activity.
  • the term host cell covers any kind of cellular system which can be engineered to generate the polypeptides and antigen binding molecules of the present invention.
  • the host cell is engineered to allow the production of an antigen binding molecule with modified glycoforms.
  • the antigen binding molecule is an antibody, antibody fragment, or fusion protein, hi certain embodiments, the host cells have been further manipulated to express increased: levels of one or more polypeptides having GnT-III activity, hi other embodiments, the host cells have been engineered to have eliminated, reduced or inhibited core ⁇ l,6- fucosyltransferase activity.
  • core al,6-fucosyltransferase activity encompasses both expression of the core ⁇ l,6-fucosyltransferase gene as well as interaction of the core ⁇ l,6-fucosyltransferase enzyme with its substrate.
  • Host cells include cultured cells, e.g., mammalian cultured cells, such as CHO cells, BHK cells, NSO cells, SP2/0 cells, YO myeloma cells, P3X63 mouse myeloma cells, PER cells, PER.C6 cells or hybridoma cells, yeast cells, insect cells, and plant cells, to name only a few, but also cells comprised within a transgenic animal, transgenic plant, or cultured plant or animal tissue.
  • mammalian cultured cells such as CHO cells, BHK cells, NSO cells, SP2/0 cells, YO myeloma cells, P3X63 mouse myeloma cells, PER cells, PER.C6 cells
  • native sequence Fc region refers to an amino acid sequence that is identical to the amino acid sequence of an Fc region commonly found in nature.
  • Exemplary native sequence human Fc regions include a native sequence human IgGl Fc region (non-A and A allotypes); native sequence human IgG2 Fc region; native sequence human IgG3 Fc region; and native sequence human IgG4 Fc region as well as naturally occurring variants thereof.
  • Other sequences are contemplated and are readily obtained from various web sites (e.g., NCBI's web site).
  • Fc receptor and FcR are used to describe a receptor that binds to an Fc region (e.g. the Fc region of an antibody or antibody fragment) of the functional equivalent of an Fc region. Portions of Fc receptors are specifically contemplated in some embodiments of the present invention.
  • the FcR is a native sequence human FcR.
  • the FcR is one which binds an IgG antibody (a gamma receptor) and includes receptors of the Fc ⁇ RI, Fc ⁇ RII, and Fc ⁇ RIII subclasses, including allelic variants and alternatively spliced forms of these receptors.
  • Fc ⁇ RII receptors include Fc ⁇ RIIa (an “activating receptor”) and Fc ⁇ RIIb (an “inhibiting receptor”), which have similar amino acid sequences that differ primarily in the cytoplasmic domains thereof.
  • Activating receptor Fc ⁇ RIIa contains an immunoreceptor tyrosine based activation motif (ITAM) in its cytoplasmic domain.
  • Inhibiting receptor Fc ⁇ RIIb contains an immunoreceptor tyrosine-based inhibition motif (ITIM) in its cytoplasmic domain.
  • ITAM immunoreceptor tyrosine based activation motif
  • ITIM immunoreceptor tyrosine-based inhibition motif
  • the term also includes the neonatal receptor, FcRn, which is responsible for the transfer of maternal IgGs to the fetus.
  • Fc receptor encompassed by the present invention is the low affinity immunoglobulin gamma Fc region receptor III-A precursor (IgG Fc receptor III-2) (Fc-gamma RHI-alpha) (Fc-gamma RIIIa) (FcRIIIa) (Fc-gamma RIII) (FcRIII) (Antigen CD16-A) (FcR-IO).
  • IgG Fc receptor III-2 low affinity immunoglobulin gamma Fc region receptor III-A precursor
  • Fc-gamma RHI-alpha Fc-gamma RIIIa
  • FcRIIIa Fc-gamma RIII
  • FcRIII Antigen CD16-A
  • FcR-IO Antigen CD16-A
  • a polypeptide variant with altered FcR binding affinity or effector function(s) is one which has either enhanced ⁇ i.e.
  • a polypeptide variant which exhibits increased binding to an FcR binds at least one FcR with better affinity than the parent polypeptide.
  • a polypeptide variant which exhibits decreased binding to an FcR binds at least one FcR with worse affinity than a parent polypeptide.
  • Such variants which display decreased binding to an FcR may possess little or no appreciable binding to an FcR, e.g., 0-20% binding to the FcR compared to a parent polypeptide.
  • a polypeptide variant which binds an FcR with increased affinity compared to a parent polypeptide is one which binds any one or more of the above identified FcRs with higher binding affinity than the parent antibody, when the amounts of polypeptide variant and parent polypeptide in a binding assay are essentially the same, and all other conditions are identical.
  • a polypeptide variant with improved FcR binding affinity may display from about 1.10 fold to about 100 fold (more typically from about 1.2 fold to about 50 fold) improvement (i.e. increase) in FcR binding affinity compared to the parent polypeptide, where FcR binding affinity is determined, for example, in an FACS-based assay or a SPR analysis (Biacore).
  • an amino acid modification refers to a change in the amino acid sequence of a given amino acid sequence.
  • exemplary modifications include, but are not limited to, an amino acid substitution, insertion, and/or deletion.
  • the amino acid modification is a substitution (e.g. in an Fc region of a parent polypeptide).
  • An amino acid modification at a specified position refers to the substitution or deletion of the specified residue, or the insertion of at least one amino acid residue adjacent the specified residue. The insertion may be N- terminal or C-terminal to the specified residue.
  • binding affinity refers to the equilibrium dissociation constant (expressed in units of concentration) associated with each Fc receptor-Fc binding interaction.
  • the binding affinity is directly related to the ratio of the kinetic off-rate (generally reported in units of inverse time, e.g. seconds.sup.-l) divided by the kinetic on-rate (generally reported in units of concentration per unit time, e.g. molar/second).
  • kinetic off-rate generally reported in units of inverse time, e.g. seconds.sup.-l
  • kinetic on-rate generally reported in units of concentration per unit time, e.g. molar/second
  • Fc-mediated cellular cytotoxicity includes antibody- dependent cellular cytotoxicity and cellular cytotoxicity mediated by a soluble Fc-fusion protein containing a human Fc-region. It is an immune mechanism leading to the lysis of "antibody-targeted cells” by "human immune effector cells”, wherein:
  • the human immune effector cells are a population of leukocytes that display Fc receptors on their surface through which they bind to the Fc-region of antibodies or of Fc- fusion proteins and perform effector functions. Such a population may include, but is not limited to, peripheral blood mononuclear cells (PBMC) and/or natural killer (NK) cells.
  • PBMC peripheral blood mononuclear cells
  • NK natural killer cells.
  • the antibody-targeted cells are cells bound by the ABMs (e.g., antibodies or Fc- fusion proteins) of the invention. In general, the antibodies or Fc fusion-proteins bind to target cells via the protein part N-terminal to the Fc region.
  • the term increased Fc-mediated cellular cytotoxicity is defined as either an increase in the number of "antibody-targeted cells” that are lysed in a given time and at a given concentration of antibody or Fc-fusion protein in the medium surrounding the target cells by the mechanism of Fc-mediated cellular cytotoxicity defined above, and/or a reduction in the concentration of antibody or Fc-fusion protein in the medium surrounding the target cells required to achieve the lysis of a given number of "antibody- targeted cells” in a given time by the mechanism of Fc-mediated cellular cytotoxicity.
  • the increase in Fc-mediated cellular cytotoxicity is relative to the cellular cytotoxicity mediated by the same antibody or Fc-fusion protein produced by the same type of host cells, using the same standard production, purification, formulation, and storage, methods which are known to those skilled in the art but which have not been produced by host cells glycoengineered to express the glycosyltransferase GnT-III by the methods described herein.
  • ADCC antibody dependent cellular cytotoxicity
  • the assay uses target cells that are known to express the target antigen recognized by the antigen binding region of the antibody;
  • PBMCs peripheral blood mononuclear cells
  • the assay is carried out according to following protocol: i) the PBMCs are isolated using standard density centrifugation procedures and are suspended at 5 x 10 6 cells/ml in RPMI cell culture medium; ii) the target cells are grown by standard tissue culture methods, harvested from the exponential growth phase with a viability higher than 90%, washed in RPMI cell culture medium, labeled with 100 micro-Curies Of 51 Cr, washed twice with cell culture medium, and resuspended in cell culture medium at a density of 10 5 cells/ml; iii) 100 ⁇ l of the final target cell suspension above are transferred to each well of a 96-well microtiter plate; iv) the antibody is serially-diluted from 4000 ng/ml to 0.04 ng/ml in cell culture medium and 50 ⁇ l of the resulting antibody solutions are added to the target cells in the 96-well microtiter plate, testing in triplicate various antibody concentrations covering the whole concentration range above; v) for the maximum release (
  • "increased ADCC” is defined as either an increase in the maximum percentage of specific lysis observed within the antibody concentration range tested above, and/or a reduction in the concentration of antibody required to achieve one half of the maximum percentage of specific lysis observed within the antibody concentration range tested above.
  • the increase in ADCC is relative to the ADCC, measured with the above assay, mediated by the same antibody, produced by the same type of host cells, using the same standard production, purification, formulation, and storage methods which are known to those skilled in the art but that has not been produced by host cells engineered to overexpress GnT-III.
  • the present invention provides polypeptides, including antigen binding molecules, having modified Fc regions, nucleic acid sequences (e.g., vectors) encoding such polypeptides, methods for generating polypeptides having modified Fc regions, and methods for using same in the treatment of various diseases and disorders.
  • the modified Fc regions of the present invention differ from the nonmodified parent Fc region by at least one amino acid modification.
  • the "parent", “starting” or “nonmodified” polypeptide preferably comprises at least a portion of an antibody Fc region, and may be prepared using techniques available in the art for generating polypeptides comprising an Fc region or portion thereof.
  • the parent polypeptide is an antibody.
  • the parent polypeptide may, however, be any other polypeptide comprising at least a portion of an Fc region (e.g. an antigen binding molecule), hi certain embodiments, a modified Fc region may be generated (e.g. according to the methods disclosed herein) and can be fused to a heterologous polypeptide of choice, such as an antibody variable domain or binding domain of a receptor or ligand.
  • a heterologous polypeptide of choice such as an antibody variable domain or binding domain of a receptor or ligand.
  • the polypeptides of the invention comprise an entire antibody comprising light and heavy chains having a modified Fc region.
  • the parent polypeptide comprises an Fc region or functional portion thereof.
  • the Fc region of the parent polypeptide will comprise a native sequence Fc region, and preferably a human native sequence Fc region.
  • the Fc region of the parent polypeptide may have one or more pre-existing amino acid sequence alterations or modifications from a native sequence Fc region.
  • the CIq binding activity of the Fc region may have been previously altered or the Fc ⁇ R binding affinity of the Fc region may have been altered.
  • the parent polypeptide Fc region is conceptual (e.g.
  • the antibody engineer may decide upon a desired modified Fc region amino acid sequence and generate a polypeptide comprising that sequence or a DNA encoding the desired modified Fc region amino acid sequence.
  • a nucleic acid encoding an Fc region of a parent polypeptide is available (e.g. commercially) and this nucleic acid sequence is altered to generate a variant nucleic acid sequence encoding the modified Fc region.
  • Polynucleotides encoding a polypeptide comprising a modified Fc region may be prepared by methods known in the art using the guidance of the present specification for particular sequences. These methods include, but are not limited to, preparation by site- directed (or oligonucleotide-mediated-) mutagenesis, PCR mutagenesis, and cassette mutagenesis of an earlier prepared nucleic acid encoding the polypeptide. Site-directed mutagenesis is a preferred method for preparing substitution variants. This technique is well known in the art (see, e.g., Carter et al. Nucleic Acids Res. 13: 4431-4443 (1985) and Kunkel et. al, Proc. Natl. Acad. Sd.
  • the starting DNA is altered by first hybridizing an oligonucleotide encoding the desired mutation to a single strand of such starting DNA. After hybridization, a DNA polymerase is used to synthesize an entire second strand, using the hybridized oligonucleotide as a primer, and using the single strand of the starting DNA as a template. Thus, the oligonucleotide encoding the desired mutation is incorporated in the resulting double- stranded DNA.
  • PCR mutagenesis is also suitable for making amino acid sequence variants of the nonmodified starting polypeptide (see, e.g., Vallette et. ah, Nuc. Acids Res. 17: 723-733 (1989), hereby incorporated by reference).
  • primers that differ slightly in sequence from the corresponding region in a template DNA can be used to generate relatively large quantities of a specific DNA fragment that differs from the template sequence only at the positions where the primers differ from the template.
  • the starting material is the plasmid (or other vector) comprising the starting polypeptide DNA to be modified.
  • the codon(s) in the starting DNA to be mutated are identified. There must be a unique restriction endonuclease site on each side of the identified mutation site(s). If no such restriction sites exist, they may be generated using the above-described oligonucleotide-mediated mutagenesis method to introduce them at appropriate locations in the starting polypeptide DNA.
  • the plasmid DNA is cut at these sites to linearize it.
  • a double-stranded oligonucleotide encoding the sequence of the DNA between the restriction sites but containing the desired mutation(s) is synthesized using standard procedures, wherein the two strands of the oligonucleotide are synthesized separately and then hybridized together using standard techniques.
  • This double-stranded oligonucleotide is referred to as the cassette.
  • This cassette is designed to have 5' and 3' ends that are compatible with the ends of the linearized plasmid, such that it can be directly ligated to the plasmid.
  • This plasmid now contains the mutated DNA sequence.
  • the desired amino acid sequence encoding a polypeptide variant can be determined, and a nucleic acid sequence encoding such amino acid sequence variant can be generated synthetically.
  • the amino acid sequence of the parent polypeptide may be modified in order to generate a variant Fc region with altered Fc receptor binding affinity or activity in vitro and/or in vivo and/or one or more altered effector functions, such as antibody-dependent cell-mediated cytotoxicity (ADCC) activity, in vitro and/or in vivo.
  • the amino acid sequence of the parent polypeptide may also be modified in order to generate a modified Fc region with altered complement binding properties and/or circulation half-life.
  • Substantial modifications in the biological properties of the Fc region may be accomplished by selecting substitutions that differ significantly in their effect on maintaining (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain.
  • Naturally occurring residues are divided into classes based on common side-chain properties:
  • hydrophobic norleucine, met, ala, val, leu, ile
  • Non-conservative substitutions will entail exchanging a member of one of these classes for a member of another class.
  • Conservative substitutions will entail exchanging a member of one of these classes for another member of the same class.
  • the modification comprises one or more of the Fc region residues identified herein (See, e.g, Table 2).
  • no more than one to about ten Fc region residues will be deleted or substituted.
  • the Fc regions herein comprising one or more amino acid modifications will preferably retain at least about 80%, and preferably at least about 90%, and most preferably at least about 95% of the parent Fc region sequence or of a native sequence human Fc region.
  • adjacent is meant within one to two amino acid residues of a Fc region residue identified herein.
  • Such Fc region variants may display enhanced or diminished FcR binding and/or effector function.
  • a co-crystal structure of a polypeptide comprising a binding region of an FcR e.g.
  • Fc ⁇ R Fc ⁇ receptor
  • FcRn Fc neonatal receptor
  • the parent polypeptide Fc region is a human Fc region, e.g. a native human IgGl (A and non-A allotypes), IgG2, IgG3, or IgG4 Fc region, including all allotypes known or discovered.
  • human Fc region e.g. a native human IgGl (A and non-A allotypes), IgG2, IgG3, or IgG4 Fc region, including all allotypes known or discovered.
  • Such regions have sequences such as those shown in SEQ ID NOS: 1-2 .
  • the parent polypeptide Fc region is a non-human Fc region.
  • Non-human Fc regions include Fc regions derived from non-human species such as, but not limited to, equine, porcine, bovine, murine, canine, feline, non-human primate, and avian subjects, e.g. a native non-human IgG Fc region, including all subclasses and allotypes known or discovered.
  • the parent polypeptide in order to generate a modified Fc region with improved effector function (e.g., ADCC), preferably has pre-existing, ADCC activity (e.g., the parent polypeptide comprises a human IgGl or human IgG3 Fc region), hi some embodiments, a modified Fc region with improved ADCC mediates ADCC substantially more effectively than an antibody with a native sequence IgGl or ⁇ gG3 Fc region.
  • ADCC activity e.g., the parent polypeptide comprises a human IgGl or human IgG3 Fc region
  • one or more amino acid modification(s) are introduced into the CH2 domain of the parent Fc region in order to generate a modified IgG Fc region with altered Fc ⁇ receptor (Fc ⁇ R) binding affinity or activity.
  • Fc ⁇ R Fc ⁇ receptor
  • the one or more amino acid modification(s) introduced into the CH2 domain of the parent Fc region occur at those positions indicated in Table 2.
  • the one or more amino acid modification(s) introduced into the CH2 domain of the parent Fc region comprises replacing the existing residue with a residue selected from the group consisting of: Tip, His, Tyr, GIu, Arg, Asp, Phe, Asn, and GIn.
  • more than one amino acid modification is introduced into the CH2 domain of the parent Fc region in order to generate a modified IgG Fc region with altered Fc ⁇ R binding affinity or activity by combining any of the individual modifications as listed in Table 2, such that a modifcation at one position can be combined with one or more additional modifications located at different positions to produce two or more modifications of the parent Fc region.
  • Fc region residues will be modified.
  • the Fc regions herein comprising one or more amino acid modifications will preferably retain at least about 80%, and preferably at least about 90%, and most preferably at least about 95% of the parent Fc region sequence or of a native sequence human Fc region.
  • one or more amino acid modification(s) introduced into the CH2 domain of the parent Fc region results in significantly reduced binding of the modified Fc region to Fc ⁇ RIIIa, e.g. those modifications listed in Table 3.
  • the one or more amino acid modification(s) introduced into the CH2 domain of the parent Fc region results in a modified IgG Fc region with only slightly reduced, unaltered, or increased affinity for Fc ⁇ RIIIa, e.g. those modifications listed in Table 4.
  • more than one amino acid modification is introduced into the CH2 domain of the parent Fc region in order to generate a modified IgG Fc region with altered Fc ⁇ R binding affinity or activity by combining any of the individual modifications as listed in Table 4, such that a modifcation at one position can be combined with one or more additional modifications located at different positions to produce any of the two or more, three or more, or four modifications of the parent Fc region listed in Table 5.
  • the more than one amino acid modification introduced into the CH2 domain of the parent Fc region involves any combination with Thr260His as listed in Table 5.
  • polypeptides of the invention having modified Fc regions may be subjected to one or more further modifications, depending on the desired or intended use of the polypeptide.
  • modifications may involve, for example, further alteration of the amino acid sequence (substitution, insertion and/or deletion of amino acid residues), fusion to heterologous polypeptide(s) and/or covalent modifications.
  • further modifications may be made prior to, simultaneously with, or following, the amino acid modification(s) disclosed above which result in an alteration of Fc receptor binding and/or effector function.
  • the starting polypeptide of particular interest in this regard is one that binds to CIq and displays complement dependent cytotoxicity (CDC).
  • Amino acid substitutions described herein may serve to alter the ability of the starting polypeptide to bind to CIq and/or modify its complement dependent cytotoxicity function (e.g. to reduce and preferably abolish these effector functions).
  • polypeptides comprising substitutions at one or more of the described positions with improved CIq binding and/or complement dependent cytotoxicity (CDC) function are contemplated herein.
  • the starting polypeptide may be unable to bind CIq and/or mediate CDC and may be modified according to the teachings herein such that it acquires these further effector functions.
  • polypeptides with pre-existing CIq binding activity, optionally further having the ability to mediate CDC may be modified such that one or both of these activities are enhanced.
  • Amino acid modifications that alter CIq and/or modify its complement dependent cytotoxicity function are described, for example, in WO00/42072, which is hereby incorporated by reference.
  • Another type of amino acid substitution serves to alter the glycosylation pattern of the polypeptide. This may be achieved, for example, by deleting one or more carbohydrate moieties found in the polypeptide, and/or adding one or more glycosylation sites that are not present in the polypeptide.
  • Glycosylation of polypeptides is typically either N-linked or O-linked. N-linked refers to the attachment of the carbohydrate moiety to the side chain of an asparagine residue.
  • the peptide sequences asparagine-X-serine and asparagine-X-threonine, where X is any amino acid except proline, are the recognition sequences for enzymatic attachment of the carbohydrate moiety to the asparagine side chain.
  • O-linked glycosylation refers to the attachment of one of the sugars N-aceylgalactosamine, galactose, or xylose to a hydroxyamino acid, most commonly serine or threonine, although 5-hydroxyproline or 5-hydroxylysine may also be used.
  • the present invention provides compositions comprising a modification of a parent polypeptide having an Fc region, wherein the modified Fc region comprises at least one surface residue amino acid modification (See, e.g., Deisenhofer, Biochemistry 20(9):2361-70 (1981), and WO00/42072, both of which are hereby incorporated by reference).
  • the present invention provides compositions comprising a modification of a parent polypeptide having an Fc region, wherein the modified Fc region comprises at least one non-surface residue amino acid modification.
  • the present invention comprises a variant of a parent polypeptide having an Fc region, wherein the variant comprises at least one surface amino acid modification and at least one non-surface amino acid modification.
  • the present invention further provides various assays for screening polypeptides of the present invention having modified Fc regions. Screening assays may be used to find or confirm useful modified Fc regions. For example, polypeptides with modified Fc regions may be screened to find variants with increased FcR binding, or effector function(s) such as ADCC, or CDC activity (e.g. increased or decreased ADCC or CDC activity). Also, modified polypeptides with amino acid modifications in non-surface residues may also be screened (e.g. a modified Fc region with a least one surface amino acid modification and one non-surface amino acid modification may be screened).
  • the assays of the present invention may be employed to find or confirm modified Fc regions that have beneficial therapeutic activity in a subject (e.g. such as a human with symptoms of an antibody or immunoadhesin responsive disease).
  • a varient of assay types may be employed to evaluate any change in a polypeptide having a modified Fc region compared to the parent polypeptide (See, screening assays provided in WO00/42072, herein incorporated by reference). Further exemplary assays are described below.
  • the polypeptides having modified Fc regions of the present invention are antigen binding molecules that essentially retain the ability to bind antigen (via an unmodified antigen binding region or modified antigen binding region) compared to the nonvariant (parent) polypeptide (e.g. the binding capability is preferably no worse than about 20 fold or no worse than about 5 fold of that of the nonvariant polypeptide).
  • the binding capability of the polypeptide variant to antigen may be determined using techniques such as fluorescence activated cell sorting (FACS) analysis or radioimmunoprecipitation (RIA), for example.
  • FACS fluorescence activated cell sorting
  • RIA radioimmunoprecipitation
  • Fc receptor (FcR) binding assays may be employed to evaluate the polypeptides with modified Fc regions of the present invention.
  • binding of Fc receptors such as Fc ⁇ RI, Fc ⁇ RIIa, Fc ⁇ RIIb, Fc ⁇ Rm, FcRn, etc.
  • Fc receptors such as Fc ⁇ RI, Fc ⁇ RIIa, Fc ⁇ RIIb, Fc ⁇ Rm, FcRn, etc.
  • an antigen binding molecule comprising a modified Fc region of the present invention may be screened in a standard ELISA assay to determine binding to an FcR.
  • a solid surface may be coated with an antigen.
  • Excess antigen may be washed, and the surface blocked.
  • the modified polypeptide (antibody) is specific for this antigen, and therefore binds to the antigen-coated surface.
  • an FcR conjugated to a label e.g. biotin
  • a molecule specific for the label on the FcR is added (e.g. avidin conjugated to an enzyme).
  • a substrate may be added in order to determine the amount of binding of the FcR to the polypeptide with the modified Fc region. The results of this assay can be compared to the ability of the parent polypeptide that lacks the modification to bind the same FcR.
  • the FcR is selected from Fc ⁇ RIIA, Fc ⁇ RIIB, and Fc ⁇ RIIIA for IgG, as these receptors (e.g. expressed recombinantly) may be successfully employed to screen the modified Fc regions of the present invention, hi fact, such binding assays with these preferred receptors unexpectedly allows the identification of useful modified Fc regions. It is unexpected that useful modified polypeptides (e.g. with greater FcR binding or effector function(s) such as ADCC or CDC) are identified in such a fashion.
  • the components for carrying out an ELISA e.g. with Fc ⁇ RIIA, Fc ⁇ RIIB, and Fc ⁇ RIIIA for IgG
  • a kit e.g. with instructions for use.
  • Useful effector cells for such assays include, but are not limited to, natural killer
  • NK peripheral blood mononuclear cells
  • PBMC peripheral blood mononuclear cells
  • ADCC activity of the polypeptides having modified Fc regions of the present invention may be assessed in vivo, e.g., in a animal model such as that disclosed in Clynes et al. PNAS (USA) 95:652-656 (1998), herein incorporated by reference).
  • modified polypeptides to bind CIq and mediate complement dependent cytotoxicity (CDC) may be assessed.
  • a CIq binding ELISA may be performed.
  • An exemplary CIq binding assay is a follows. Assay plates may be coated overnight at 4 0 C with modified polypeptide of the invention or parental polypeptide (control) in coating buffer. The plates may then be washed and blocked. Following washing, an aliquot of human CIq may be added to each well and incubated for 2 hrs at room temperature. Following a further wash, 100 ⁇ l of a sheep anti- complement CIq peroxidase-conjugated antibody may be added to each well and incubated for 1 hour at room temperature.
  • the plate may again be washed with wash buffer and 100 ⁇ l of substrate buffer containing OPD (O-phenylenediamine dihydrochloride (Sigma)) may be added to each well.
  • OPD O-phenylenediamine dihydrochloride (Sigma)
  • the oxidation reaction observed by the appearance of a yellow color, may be allowed to proceed for an optimized time (2-60 minutes) and stopped by the addition of 100 ⁇ l of 4.5 N H 2 SO 4 .
  • the absorbance may then be read at 492 nm and the background absorbance at 405 nm subtracted from this value.
  • the modified Fc regions of the present invention may also be screened for complement activation.
  • a complement dependent cytotoxicity (CDC) assay may be performed (See, e.g. Gazzano-Santoro et ah, J. Immunol. Methods, 202:163 (1996), herein incorporated by reference).
  • CDC complement dependent cytotoxicity
  • various concentrations of the modified polypeptide of the invention and human complement may be diluted with buffer.
  • Cells which express the antigen to which the polypeptide variant binds may be diluted to a density of ⁇ lxl0 6 cells/ml.
  • Polypeptide variant, diluted human complement and cells expressing the antigen may be added to a flat bottom tissue culture 96 well plate and allowed to incubate for 2 hours at 37° C and 5% CO 2 to facilitate complement mediated cell lysis. 50 ⁇ l of alamar blue (Accumed International) may then be added to each well and incubated overnight at 37° C. The absorbance may be measured using a 96-well fiuorimeter with excitation at 530 nm and emission at 590 nm. The results may be expressed in relative fluorescence units (RFU). The sample concentrations may be computed from a standard curve and the percent activity as compared to nonvariant polypeptide may be reported for the polypeptide variant of interest.
  • RNU relative fluorescence units
  • the modified polypeptide has a higher binding affinity for human CIq than the parent polypeptide.
  • a variant may display, for example, about two-fold or more, and preferably about five-fold or more improvement in human CIq binding compared to the parent polypeptide (e.g. at the IC50 values for these two molecules).
  • human CIq binding may be about two-fold to about 500-fold, and preferably from about two-fold or from about five-fold to about 1000-fold improved compared to the parent polypeptide.
  • variants are found that exhibit 2-fold, 25-fold,
  • the modified Fc region polypeptide does not bind CIq (e.g., 10 ⁇ g/ml of the modified polypeptide displays about 100 fold or more reduction in CIq binding compared to 10 ⁇ g/ml of the control antibody).
  • the modified polypeptides of the present invention do no activate complement.
  • a modified polypeptide displays about 0-10% CDC activity in this assay compared to a control antibody having a nonmodified IgGl Fc region.
  • the variant does not appear to have any CDC activity (e.g. above background) in the above CDC assay.
  • the modified polypeptides of the present invention are found to have enhanced CDC compared to a parent polypeptide [e.g., displaying about two-fold to about 100-fold (or greater) improvement in CDC activity in vitro or in vivo when the IC50 values are compared].
  • the polypeptides having modified Fc regions of the present invention may also be screened in vivo. Any type of in vivo assay may be employed. A particular example of one type of assay is provided below. This exemplary assay allows for preclinical evaluation of modified Fc regions in vivo.
  • a modified polypeptide to be tested may be incorporated into the Fc region of a particular antibody known to have some activity. For example, a modification may be incorporated into the Fc region of an anti-CD20 IgG by mutagenesis. This allows a parental IgG and Fc variant IgG to be compared directly with RITUXAN (known to promote tumor regression).
  • the preclinical evaluation may be done in 2 phases (a pharmacokinetic and pharmacodynamic phase).
  • the goal of the Phase I pharmacokinetic studies is to determine if there are differences in the clearance rate between an Fc variant IgG and the antibody with known in vivo activity (e.g. RITUXAN). Differences in clearance rate may cause differences in the steady-state level of IgG in serum. As such, if differences in steady-state concentrations are detected these should be normalized to enable accurate comparisons to be made.
  • the goal of the Phase II pharmacodynamic studies is to determine the effect of the Fc mutations upon, in this case, tumor growth. Previous studies with RITUXAN used a single dose which completely inhibited tumor growth. Because this does not allow quantitative differences to be measured, a dose range should be employed.
  • Phase I pharmacokinetic comparison of a polypeptide having a modified Fc region of the present invention, the nonmodified (e.g., wild type) parental Fc, and RITUXAN may be performed in the following manner.
  • 40 ⁇ g per animal may be injected intravenously and the plasma level of the IgG quantitated at 0, 0.25, 0.5, 1, 24, 48, 168, and 336 hrs.
  • the data may be fitted, for example, using a pharmacokinetic program (WinNonLin) using a zero lag two compartment pharmacokinetic model to obtain the clearance rate.
  • WinNonLin pharmacokinetic program
  • Pharmacokinetic experiments may be performed in non-tumor bearing mice with, for example, a minimum of 5 mice per time point.
  • An animal model may be employed for the next phase in the following manner.
  • the right flank of CB 17-SCID mice may be implanted with 106 Raji cells subcutaneously.
  • Intravenous bolus of the polypeptide with modified Fc region ⁇ , the polypeptide with the parent (e.g.,wild type) Fc, and RITUXAN may be commenced immediately after implantation and continued until the tumor size is greater than 2 cm in diameter.
  • a plot of tumor volume versus time will give the tumor growth rate for the pharmakodynamic calculation.
  • a minimum of about 10 animals per group should be used.
  • Phase II pharmacodynamic comparison of the polypeptide with modified Fc region of the invention, the parental (e.g., wild type) Fc, and RITUXAN may be performed in the following manner. Based on published data, RITUXAN at 10 ⁇ g/g weekly completely inhibited tumor growth in vivo (dynes et at, Nat. Med. 2000 April;6(4):443-6, 2000, herein incorporated by reference). Therefore, a weekly dose range of 10 ⁇ g/g, 5 ⁇ g/g, 1 ⁇ g/g, 0.5 ⁇ g/g, and 0 ⁇ g/g may be tested.
  • the steady state plasma level at which tumor growth rate is inhibited by 50% may be graphically determined by the relationship between steady state plasma level and effectiveness.
  • the steady state plasma level may be calculated as described above. If necessary, T may be adjusted accordingly for each modified Fc region polypeptide and the Fc wild type depending on their pharmacokinetic properties to achieve comparable steady state plasma level as RITUXAN.
  • Statistical improved pharmakodynamic values of the modified polypeptide in comparison to the parental polypeptide (e.g. Fc wild type) and RITUXAN will generally indicate that the modified polypeptide confers improved activity in vivo.
  • the modified Fc regions of the present invention are screened such that variants that are useful for therapeutic use in at least two species are identified.
  • variants are referred to herein as "dual-species improved variants," and are particularly useful for identifying variants that are therapeutic in humans, and also demonstrate (or are likely to demonstrate) efficacy in an animal model.
  • the present invention provides methods for identifying variants that have a strong chance of being approved for human clinical testing since animal model data will likely support any human testing applications made to governmental regulatory agencies (e.g. U.S. Food and Drug Administration).
  • dual-species improved modified polypeptides are identified by first performing an ADCC assay using human effector cells; to find improved modified polypeptides, and then performing a second ADCC assay using mouse, rat, or non-human primate effector cells to identify a sub-set of the improved modified polypeptides that are dual-species improved modified polypeptides.
  • the present invention provides methods for identifying dual-species improved modified polypeptides, comprising: a) providing: i) target cells, ii) a composition comprising a candidate modified polypeptide of a parent polypeptide having at least a portion of an Fc region, wherein the candidate modified polypeptide comprises at least one amino acid modification in the Fc region, and wherein the candidate modified polypeptide mediates target cell cytotoxicity in the presence of a first species (e.g.
  • the method further comprises step e) determining if the candidate modified polypeptide mediates target cell cytotoxicity in the presence of the second species effector cells more effectively than the parent polypeptide, hi some embodiments, the method further comprises step f) identifying a candidate modified polypeptide as a dual-species improved modified polypeptide that mediates target cell cytotoxicity in the presence of the second species effector cells more effectively than the parent polypeptide.
  • the dual-species modified polypeptides identified are then
  • dual-species improved modified polypeptides are identified by performing any of the assays above using human components (e.g. human cells, human Fc receptors, etc.) to identify improved polypeptides having modified Fc regions, and then running the same assay (or a different assay) with non-human animal components (e.g. mouse cells, mouse Fc receptors, etc.).
  • human components e.g. human cells, human Fc receptors, etc.
  • non-human animal components e.g. mouse cells, mouse Fc receptors, etc.
  • a sub-set of modified polypeptides that perform well according to a given criteria in both human based assays and a second species based assays can be identified.
  • An exemplary process for identifying dual-species improved polypeptides having modified Fc regions of the invention is a follows. First, a nucleic acid sequence encoding at least a portion of an IgG Fc region is modified such that the amino acid sequence expressed has at least one amino acid change, thereby generating a modified Fc region. This expressed IgG variant is then captured via antigen on an assay plate. Next, the captured variant is screened for soluble human Fc ⁇ RIII binding using ELISA. If the variant demonstrates improved or comparable (compared to a non-mutated parental Fc region) Fc ⁇ RIII binding, then the variant is screened for human Fc ⁇ RIII binding using ELISA. The relative specificity ratio for the variant may then be calculated.
  • an ADCC assay is performed with the variant using human PBMCs or a subset (NK cells or macrophages, for example). If enhanced ADCC activity is found, then the variant is screened in a second ADCC assay using mouse or rat PBMCs. Alternatively, or in addition, an assay can be performed with the variant for binding to cloned rodent receptors or cell lines. Finally, if the variant is found to be improved in the second assay, making it a dual-improved variant, then the variant is screened in vivo in mice or rats.
  • the variant Fc regions of the present invention may be part of larger molecules, preferably antigen binding molecules (ABMs).
  • the larger molecules may be, for example, monoclonal antibodies, polyclonal antibodies, chimeric antibodies, humanized antibodies, bispecific antibodies, immunoadliesins, etc. As such, it is evident that there is a broad range of applications for the modified Fc regions of the present invention.
  • the antigen binding molecules comprising the modified Fc regions of the present invention may be optimized for a variety of properties. Properties that may be optimized include, but are not limited to, enhanced or reduced affinity for an Fc ⁇ R. hi a preferred embodiment, the modified Fc regions of the present invention are optimized to possess enhanced affinity for a human activating Fc ⁇ R, preferably FcRI, Fc ⁇ RIIa, Fc ⁇ RIIc, Fc ⁇ RIIIa, and Fc ⁇ RIIIb, most preferably Fc ⁇ RIIIa. In an alternately preferred embodiment, the modified Fc regions are optimized to possess reduced affinity for the human inhibitory receptor Fc ⁇ RIIb.
  • the ABMs of the invention provide antibodies and Fc fusions with enhanced therapeutic properties in humans, for example enhanced effector function and greater anti-cancer potency, hi an alternate embodiment, the modified Fc regions of the present invention are optimized to have reduced or ablated affinity for a human Fc ⁇ R, including but not limited to Fc ⁇ RI, Fc ⁇ RIIa, Fc ⁇ RIIb, or Fc ⁇ RIIc.
  • Fc ⁇ RI Fc ⁇ RIIa
  • Fc ⁇ RIIb Fc ⁇ RIIc
  • Fc ⁇ RIIc Fc ⁇ RIIc
  • These ABMs of the invention are anticipated to provide antibodies and Fc fusions with enhanced therapeutic properties in humans, for example reduced effector function and reduced toxicity.
  • Preferred embodiments comprise optimization of Fc binding to a human Fc ⁇ R; however, in alternate embodiments, the Fc variants of the present invention possess enhanced or reduced affinity for Fc ⁇ Rs from nonhuman organisms, including but not limited to mice, rats, rabbits, and monkeys.
  • Fc variants that are optimized for binding to a nonhuman Fc ⁇ R may find use in experimentation.
  • mouse models are available for a variety of diseases that enable testing of properties such as efficacy, toxicity, and pharmacokinetics for a given drug candidate.
  • cancer cells can be grafted or injected into mice to mimic a human cancer, a process referred to as xenografting.
  • Testing of antibodies or Fc fusions that comprise modified Fc regions that are optimized for one or more mouse Fc ⁇ Rs may provide valuable information with regard to the efficacy of the antibody or Fc fusion, its mechanism of action, and the like.
  • the modified Fc regions of the present invention may be derived from parent Fc polypeptides that are themselves from a wide range of sources.
  • the parent Fc polypeptide may be substantially encoded by one or more Fc genes from any organism, including but not limited to humans, mice, rats, rabbits, camels, llamas, dromedaries, monkeys, preferably mammals, and most preferably humans and mice.
  • the parent Fc polypeptide comprises an antibody, referred to as the parent antibody.
  • the parent antibody may be fully human, obtained for example using transgenic mice (Bruggemann et al., 1997, Curr Opin Biotechnol ⁇ :455-458) or human antibody libraries coupled with selection methods (Griffiths et al., 1998, Curr Opin Biotechnol 9:102-108).
  • the parent antibody need not be naturally occurring.
  • the parent antibody may be an engineered antibody, including but not limited to chimeric antibodies and humanized antibodies (Clark, 2000, Immunol Today 27:397-402).
  • the parent antibody may be an engineered variant of an antibody that is substantially encoded by one, or more natural antibody genes.
  • the parent antibody has been affinity matured, as is known in the art.
  • the antibody has been modified in some other way, for example as described in U.S. Ser. No. 10/339,788, filed on Mar. 3, 2003.
  • the modified Fc regions of the present invention may be substantially encoded by immunoglobulin genes belonging to any of the antibody classes.
  • the modified Fc regions of the present invention find use in antibodies or Fc fusions that comprise sequences belonging to the IgG class of antibodies, including IgGl, IgG2, IgG3, or IgG4.
  • the modified Fc regions of the present invention find use in antibodies or Fc fusions that comprise sequences belonging to the IgA (including subclasses IgAl and IgA2), IgD, IgE, IgG, or IgM classes of antibodies.
  • the modified Fc regions of the present invention may comprise more than one protein chain. That is, the present invention may find use in an antibody or Fc fusion that is a monomer or an oligomer, including a homo- or hetero-oligomer.
  • modified Fc regions of the present invention may be combined with other Fc modifications, including but not limited to modifications that alter effector function or interaction with one or more Fc ligands. Such combination may provide additive, synergistic, or novel properties in the ABMs of the invention.
  • the modified Fc regions of the present invention may be combined with other known Fc modifications (Duncan et ah, 1988, Nature 352:563-564; Lund et ah, 1991, J Immunol 147:2657-2662; Lund et ah, 1992, MoI Immunol 29:53-59; Alegre et ah, 1994, Transplantation 57:1537-1543; Hutchins et ah, 1995, Proc Natl Acad Sci USA 02:11980- 11984; Jefferis et ah, 1995, Immunol Left 44:111-117; Lund et ah, 1995, Faseb ./9:115- 119; Jefferis et ah, 1996, Immunol Left 54:101-104; Lund et ah, 1996, J Immunol 157:4963-4969; Armour et ah, 1999, Eur J Immunol 29:26
  • Virtually any antigen may be targeted by ABMs comprising the modified Fc regions of the invention, including but not limited to the following list of proteins, subunits, domains, motifs, and epitopes belonging to the following list of proteins: CD2; CD3, CD3E, CD4, CDIl, CDlIa, CD14, CD16, CD18, CD19, CD20, CD22, CD23, CD25, CD28, CD29, CD30, CD32, CD33 (p67 protein), CD38, CD40, CD40L, CD52, CD54, CD56, CD80, CD147, GD3, IL-I, IL-IR, IL-2, IL-2R, IL-4, IL-5, IL-6, IL-6R, IL- 8, IL-12, IL-15, IL-18, IL-23, interferon ⁇ , interferon ⁇ , interferon ⁇ ; TNF- ⁇ , TNF ⁇ 2, TNFc, TNF ⁇ , TNF-RI, TNF-RII, FasL
  • targets refers not only to specific proteins and biomolecules, but the biochemical pathway or pathways that comprise them.
  • CTLA-4 as a target antigen implies that the ligands and receptors that make up the T cell co-stimulatory pathway, including CTLA-4, B7-1, B7-2, CD28, and any other undiscovered ligands or receptors that bind these proteins, are also targets.
  • target as used herein refers not only to a specific biomolecule, but the set of proteins that interact with said target and the members of the biochemical pathway to which said target belongs.
  • any of the aforementioned target antigens, the ligands or receptors that bind them, or other members of their corresponding biochemical pathway may be operably linked to the Fc variants of the present invention in order to generate an Fc fusion.
  • an Fc fusion that targets EGFR could be constructed by operably linking an Fc variant to EGF, TGF ⁇ , or any other ligand, discovered or undiscovered, that binds EGFR.
  • a modified Fc region of the present invention could be operably linked to EGFR in order to generate an Fc fusion that binds EGF, TGF ⁇ , or any other ligand,- discovered or undiscovered, that binds EGFR.
  • any polypeptide whether a ligand, receptor, or some other protein or protein domain, including but not limited to the aforementioned targets and the proteins that compose their corresponding biochemical pathways, may be operably linked to the Fc variants of the present invention to develop an Fc fusion.
  • a number of antibodies and Fc fusions that are approved for use, in clinical trials, or in development may benefit from the modified Fc regions of the present invention.
  • Said antibodies and Fc fusions are herein referred to as "clinical products and candidates.”
  • the Fc variants of the present invention may find use in a range of clinical products and candidates.
  • a number of antibodies that target CD20 may benefit from the modified Fc regions of the present invention.
  • the modified Fc regions of the present invention may find use in an antibody that is substantially similar to rituxirnab (Rituxan®, IDEC/Genentech/Roche) (see for example U.S. Pat. No. 5,736,137), a chimeric anti-CD20 antibody approved to treat Non- Hodgkin's lymphoma; HuMax-CD20, an anti-CD20 currently being developed by Genmab; an anti-CD20 antibody described in U.S. Pat. No. 5,500,362;: AME-I33 (Applied Molecular Evolution); hA20 (Immunomedics, Inc.); and HumaLYM (fritracel).
  • rituxirnab Rostuxan®, IDEC/Genentech/Roche
  • a number of antibodies that target members of the family of epidermal growth factor receptors may benefit from the Fc variants of the present invention.
  • the Fc variants of the present invention may find use in an antibody that is substantially similar to trastuzumab (Herceptin®, Genentech) (see for example U.S. Pat. No. 5,677,171), a humanized anti-Her2/neu antibody approved to treat breast cancer; pertuzumab (rhuMab- 2C4, Omnitarg.TM.), currently being developed by Genentech; an anti-Her2 antibody described in U.S. Pat. No.
  • the modified Fc regions of the present invention may find use in alemtuzumab (Campath®, Millenium), a humanized monoclonal antibody currently approved for treatment of B-cell chronic lymphocytic leukemia.
  • the modified Fc regions may find use in a variety of antibodies or Fc fusions that are substantially similar to other clinical products and candidates, including but not limited to muromonab-CD3 (Orthoclone OKT3®)), an anti-CD3 antibody developed by Ortho Biotech/Johnson & Johnson, ibritumomab tiuxetan (Zevalin®), an anti-CD20 antibody developed by IDEC/Schering AG, gemtuzumab ozogamicin (Mylotarg®), an anti-CD33 (p67 protein) antibody developed by Celltech/Wyeth, alefacept (Amevive®), an anti-LFA-3 Fc fusion developed by Biogen), abciximab (ReoPro®)), developed by Centocor/Lilly, basiliximab (Simulect®.)), developed by Novartis, palivizumab (Synagis®)), developed by Medlmmune, infliximab (Remicade®)),
  • an anti-Blys antibody being developed by Cambridge Antibody Technology and Human Genome Sciences Inc., TRAIL-RImAb, an anti-TRAIL-Rl antibody being developed by Cambridge Antibody Technology and Human Genome Sciences, Inc., Avastin®(bevacizumab, rhuMAb- VEGF), an anti-VEGF antibody being developed by Genentech, an anti-HER receptor family antibody being developed by Genentech, Anti-Tissue Factor (ATF), an anti-Tissue Factor antibody being developed by Genentech, Xolair® (Omalizumab), an anti-IgE antibody being developed by Genentech, Raptiva®(Efalizumab), an anti-CDlla antibody being developed by Genentech and Xoma, MLN-02 Antibody (formerly LDP-02), being developed by Genentech and Millenium Pharmaceuticals, HuMax CD4, an anti-CD4 antibody being developed by Genmab, HuMax-IL 15, an anti-IL15 antibody being developed by Genmab
  • modified Fc regions to the aforementioned antibody and Fc fusion clinical products and candidates is not meant to be constrained to their precise composition.
  • the modified Fc regions of the present invention may be incorporated into the aforementioned clinical candidates and products, or into antibodies and Fc fusions that are substantially similar to them.
  • the modified Fc regions of the present invention may be incorporated into versions of the aforementioned clinical candidates and products that are humanized, affinity matured, engineered, or modified in some ⁇ other way.
  • the entire polypeptide of the aforementioned clinical products and candidates need not be used to construct a new antibody or Fc fusion that incorporates the modified Fc region of the present invention; for example only the variable region of a clinical product or candidate antibody, a substantially similar variable region, or a humanized, affinity matured, engineered, or modified version of the variable region may be used.
  • the modified Fc region of the present invention may find use in an antibody or Fc fusion that binds to the same epitope, antigen, ligand, or receptor as one of the aforementioned clinical products and candidates.
  • the modified Fc regions of the present invention may find use in a wide range of antibody and Fc fusion products.
  • the ABM of the present invention is a therapeutic, a diagnostic, or a research reagent, preferably a therapeutic.
  • Diseases and disorders capable of being treated or ameliorated by the ABM of the invention include, but are not limited to, autoimmune diseases, immunological diseases, infectious diseases, inflammatory diseases, neurological diseases, and oncological and neoplastic diseases including cancer.
  • cancer and “cancerous” herein refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth.
  • cancers include but are not limited to carcinoma, lymphoma, blastoma, sarcoma (including liposarcoma), neuroendocrine tumors, mesothelioma, schwanoma, meningioma, adenocarcinoma, melanoma, and leukemia or lymphoid malignancies. More particular examples of such cancers include squamous cell cancer (e.g.
  • lung cancer including small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, testicular cancer, esophagael cancer, tumors of the biliary tract, as well as head and neck cancer.
  • lung cancer including small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer,
  • the Fc variants of the present invention may be used to treat conditions including but not limited to congestive heart failure (CHF), vasculitis, rosecea, acne, eczema, myocarditis and other conditions of the myocardium, systemic lupus erythematosus, diabetes, spondylopathies, synovial fibroblasts, and bone marrow stroma; bone loss; Paget's disease, osteoclastoma; multiple myeloma; breast cancer; disuse osteopenia; malnutrition, periodontal disease, Gaucher's disease, Langerhans' cell histiocytosis, spinal cord injury, acute septic arthritis, osteomalacia, Cushing's syndrome, monoostotic fibrous dysplasia, polyostotic fibrous dysplasia, periodontal reconstruction, and bone fractures; sarcoidosis; multiple myeloma; osteolytic bone cancers, breast cancer, lung cancer, kidney cancer and rectal cancer; bone metastas
  • an ABM comprising a modified Fc region of the present invention is administered to a patient having a disease involving inappropriate expression of a protein.
  • this is meant to include diseases and disorders characterized by aberrant proteins, due for example to alterations in the amount of a protein present, the presence of a mutant protein, or both.
  • An overabundance may be due to any cause, including but not limited to overexpression at the molecular level, prolonged or accumulated appearance at the site of action, or increased activity of a protein relative to normal. Included within this definition are diseases and disorders characterized by a reduction of a protein.
  • This reduction may be due to any cause, including but not limited to reduced expression at the molecular level, shortened or reduced appearance at the site of action, mutant forms of a protein, or decreased activity of a protein relative to normal.
  • Such an overabundance or reduction of a protein can be measured relative to normal expression, appearance, or activity of a protein, and said measurement may play an important role in the development and/or clinical testing of the ABMs of the present invention.
  • the present invention provides engineering methods that may be used to generate
  • Fc variants A principal obstacle that has hindered previous attempts at Fc engineering is that only random attempts at modification have been possible, due in part to the inefficiency of engineering strategies and methods, and to the low-throughput nature of antibody production and screening.
  • the present invention describes engineering methods that overcome these shortcomings.
  • a variety of design strategies, computational screening methods, library generation methods, and experimental production and screening methods are contemplated. These strategies, approaches, techniques, and methods may be applied individually or in various combinations to engineer optimized Fc variants.
  • Fc with some Fc ligand is altered by engineering amino acid modifications at the interface between Fc and said Fc ligand.
  • Fc ligands herein may include but are not limited to Fc ⁇ Rs, CIq, FcRn, protein A or G, and the like.
  • variants can be engineered that sample new interface conformations, some of which may improve binding to the Fc ligand, some of which may reduce Fc ligand binding, and some of which may have other favorable properties.
  • Such new interface conformations could be the result of, for example, direct interaction with Fc ligand residues that form the interface, or indirect effects caused by the amino acid modifications such as perturbation of side chain or backbone conformations.
  • Variable positions may be chosen as any positions that are believed to play an important role in determining the conformation of the interface.
  • variable positions may be chosen as the set of residues that are within a certain distance, for example 5 Angstroms, preferably between 1 and 10 Angstroms, of any residue that makes direct contact with the Fc ligand.
  • An additional design strategy for generating Fc variants is provided in which the conformation of the Fc carbohydrate at N297 is optimized. Optimization as used in this context is meant to include conformational and compositional changes in the N297 carbohydrate that result in a desired property, for example increased or reduced affinity for an Fc ⁇ R.
  • Another design strategy for generating Fc variants is provided in which the angle between the C ⁇ 2 and C ⁇ 3 domains is optimized. Optimization as used in this context is meant to describe conformational changes in the C ⁇ 2-C ⁇ 3 domain angle that result in a desired property, for example increased or reduced affinity for an Fc ⁇ R.
  • This angle is an important determinant of Fc/Fc ⁇ R affinity (Radaev et al, 2001, J Biol Chem 27(5:16478- 16483), and a number of mutations distal to the Fc/Fc ⁇ R interface affect binding potentially by modulating it (Shields et al, J Biol Chem 27(5:6591-6604 (2001)).
  • Fc is reengineered to eliminate the structural and functional dependence on glycosylation.
  • This design strategy involves the optimization of Fc structure, stability, solubility, and/or Fc function (for example affinity of Fc for one or more Fc ligands) in the absence of the N297 carbohydrate.
  • positions that are exposed to solvent in the absence of glycosylation are engineered such that they are stable, structurally consistent with Fc structure, and have no tendency to aggregate.
  • the C ⁇ 2 is the only unpaired Ig domain in the antibody.
  • N297 carbohydrate covers up the exposed hydrophobic patch that would normally be the interface for a protein-protein interaction with another Ig domain, maintaining the stability and structural integrity of Fc and keeping the C ⁇ 2 domains from aggregating across the central axis.
  • Approaches for optimizing aglycosylated Fc may involve but are not limited to designing amino acid modifications that enhance aglycoslated Fc stability and/or solubility by incorporating polar and/or charged residues that face inward towards the C ⁇ 2-C ⁇ 2 dimer axis, and by designing amino acid modifications that directly enhance the aglycosylated Fc/Fc ⁇ R interface or the interface of aglycosylated Fc with some other Fc ligand.
  • An additional design strategy for engineering Fc variants is provided in which the conformation of the C ⁇ 2 domain is optimized. Optimization as used in this context is meant to describe conformational changes in the C ⁇ 2 domain angle that result in a desired property, for example increased or reduced affinity for an Fc ⁇ R.
  • a quality diversity of variants can be engineered that sample new C ⁇ 2 conformations, some of which may achieve the design goal.
  • Such new C ⁇ 2 conformations could be the result of, for example, alternate backbone conformations that are sampled by the variant.
  • Variable positions may be chosen as any positions that are believed to play an important role in determining C ⁇ 2 structure, stability, solubility, flexibility, function, and the like.
  • C ⁇ 2 hydrophobic core residues that is C ⁇ 2 residues that are partially or fully sequestered from solvent, may be reengineered.
  • noncore residues may be considered, or residues that are deemed important for determining backbone structure, stability, or flexibility.
  • An additional design strategy for Fc optimization is provided in which binding to an Fc ⁇ R, complement, or some other Fc ligand is altered by modifications that modulate the electrostatic interaction between Fc and said Fc ligand.
  • modifications may be thought of as optimization of the global electrostatic character of Fc, and include replacement of neutral amino acids with a charged amino acid, replacement of a charged amino acid with a neutral amino acid, or replacement of a charged amino acid with an amino acid of opposite charge (i.e. charge reversal).
  • Such modifications may be used to effect changes in binding affinity between an Fc and one or more Fc ligands, for example Fc.gamma.Rs.
  • positions at which electrostatic substitutions might affect binding are selected using one of a variety of well known methods for calculation of electrostatic potentials. Ih the simplest embodiment, Coulomb's law is used to generate electrostatic potentials as a function of the position in the protein. Additional embodiments include the use of Debye-Huckel scaling to account for ionic strength effects, and more sophisticated embodiments such as Poisson-Boltzmann calculations. Such electrostatic calculations may highlight positions and suggest specific amino acid modifications to achieve the design goal. In some cases, these substitutions may be anticipated to variably affect binding to different Fc ligands, for example to enhance binding to activating Fc ⁇ Rs while decreasing binding affinity to inhibitory Fc ⁇ Rs: ,
  • the chimeric ABM of the present invention is a humanized antibody.
  • Methods for humanizing non-human antibodies are known in the art.
  • humanized ABMs of the present invention can be prepared according to the methods of U.S. Pat. No. 5,225,539 to Winter; U.S. Pat. No. 6,180,370 to Queen et al; U.S. Pat. No. 6,632,927 to Adair et al; U.S. Pat. Appl. Pub. No. 2003/0039649 to Foote; U.S. Pat. Appl. Pub. No. 2004/0044187 to Sato et al; or U.S. Pat. Appl. Pub. No.
  • a humanized antibody has one or more amino acid residues introduced into it from a source which is non-human. These non-human amino acid residues are often referred to as "import" residues, which are typically taken from an “import” variable domain.
  • Humanization can be essentially performed following the method of Winter and co-workers (Jones et ah, Nature, 321:522- 525 (1986); Riechmann et ah, Nature, 332:323-327 (1988); Verhoeyen et ah, Science, 239:1534-1536 (1988)), by substituting hypervariable region sequences for the corresponding sequences of a human antibody. Accordingly, such "humanized" antibodies are chimeric antibodies (U.S. Pat. No. 4,816,567) wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species.
  • humanized antibodies are typically human antibodies in which some hypervariable region residues and possibly some FR residues are substituted by residues from analogous sites in rodent antibodies.
  • the subject humanized antibodies will generally comprise constant regions of human immunoglobulins, such as IgGl.
  • the choice of human variable domains, both light and heavy, to be used in making the humanized antibodies is very important to reduce antigenicity.
  • the sequence of the variable domain of a rodent antibody is screened against the entire library of known human variable-domain sequences. The human sequence which is closest to that of the rodent is then accepted as the human framework region (FR) for the humanized antibody (Sims et ah, J.
  • FR human framework region
  • Another method of selecting the human framework sequence is to compare the sequence of each individual subregion of the full rodent framework (i.e., FRl, FR2, FR3, and FR4) or some combination of the individual subregions (e.g., FRl and FR2) against a library of known human variable region sequences that correspond to that framework subregion (e.g., as determined by Kabat numbering), and choose the human sequence for each subregion or combination that is the closest to that of the rodent (Leung U.S. Patent Application Publication No. 2003/0040606 Al, published Feb.
  • Another method uses a particular framework region derived from the consensus sequence of all human antibodies of a particular subgroup of light or heavy chains. The same framework may be used for several different humanized antibodies (Carter et ah, Proc. Natl. Acad. ScL USA, ⁇ P:4285 (1992); Presta et ah, J. Immunol., 151:2623 (1993)) (the entire contents of each of which are hereby incorporated by reference). [0152] It is further important that antibodies be humanized with retention of high affinity for the antigen and other favorable biological properties.
  • humanized antibodies are prepared by a process of analysis of the parental sequences and various conceptual humanized products using three-dimensional models of the parental and humanized sequences.
  • Three-dimensional immunoglobulin models can be generated using computer programs familiar to those skilled in the art (e.g. InsightII, accelrys inc (former MSI), or at http://swissmodel.expasy.org/ described by Schwede et al., Nucleic Acids Res. 2003 (13):3381-3385). Inspection of these models permits analysis of the likely role of the residues in the functioning of the candidate immunoglobulin sequence, i.e., the analysis of residues that influence the ability of the candidate immunoglobulin to bind its antigen.
  • FR residues can be selected and combined from the recipient and import sequences so that the desired antibody characteristic, such as maintained affinity for the target antigen(s), is achieved»In ! general, the hypervariable region residues are directly and most substantially involved in influencing antigen binding.
  • the ABMs of the present invention comprise a modified human Fc region, hi a specific embodiment, the human constant region is IgGl, as set forth in SEQ ID NOs 1 and 2, and set forth below:
  • variants and isoforms of the native human Fc region are also encompassed by the present invention.
  • variant Fc regions suitable for use in the present invention can be produced according to the methods taught in U.S. Pat. No. 6,737,056 to Presta (Fc region variants with altered effector function due to one or more amino acid modifications); or in U.S. Pat. Appl. Nos. 60/439,498; 60/456,041; 60/514,549; or WO 2004/063351 (variant Fc regions with increased binding affinity due to amino acid modification.); or in U.S. Pat. No. 10/672,280 or WO 2004/099249 (Fc variants with altered binding to Fc ⁇ R due to amino acid modification), the contents of each of which are incorporated herein by reference in their entirety.
  • the antigen binding molecules of the present invention are engineered to have enhanced binding affinity according to, for example, the methods disclosed in U.S. Pat. Appl. Pub. No. 2004/0132066 to Balint et ah, the entire contents of which are hereby incorporated by reference.
  • the antigen binding molecule of the present invention is conjugated to an additional moiety, such as a radiolabel or a toxin.
  • an additional moiety such as a radiolabel or a toxin.
  • ABMs can be produced by numerous methods that are well known in the art.
  • 131 iodine is a well known radionuclide used for targeted immunotherapy.
  • the clinical usefulness of 131 iodine can be limited by several factors including: eight-day physical half-life; dehalogenation of iodinated antibody both in the blood and at tumor sites; and emission characteristics (eg, large gamma component) which can be suboptimal for localized dose deposition in tumor.
  • 90 Yttrium provides several benefits for utilization in radioimmunotherapeutic applications: the 64 hour half-life of 90 yttrium is long enough to allow antibody accumulation by tumor and, unlike eg, 131 iodine, 90 yttrium is a pure beta emitter of high energy with no accompanying gamma irradiation in its decay, with a range in tissue of 100 to 1000 cell diameters. Furthermore, the minimal amount of penetrating radiation allows for outpatient administration of 90 yttrium.-labeled antibodies. Additionally, internalization of labeled antibody is not required for cell killing, and the local emission of ionizing radiation should be lethal for adjacent tumor cells lacking the target antigen.
  • radiolabeled antibodies therapy therewith can also occur using a single therapy treatment or using multiple treatments. Because of the radionuclide component, it is preferred that prior to treatment, peripheral stem cells (“PSC”) or bone marrow (“BM”) be “harvested” for patients experiencing potentially fatal bone marrow toxicity resulting from radiation. BM and/or PSC are harvested using standard techniques, and then purged and frozen for possible reinfusion.
  • PSC peripheral stem cells
  • BM bone marrow
  • a diagnostic dosimetry study using a diagnostic labeled antibody be conducted on the patient, a purpose of which is to ensure that the therapeutically labeled antibody (eg, using 90 yttrmrn) will not become unnecessarily "concentrated” in any normal organ or tissue.
  • the present invention is directed to an isolated polynucleotide comprising a sequence that encodes a polypeptide of the invention.
  • the invention is further directed to an isolated nucleic acid comprising a sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to a nucleotide sequence of the invention.
  • the invention is directed to an isolated nucleic acid comprising a sequence that encodes a polypeptide having an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to an amino acid sequence of the invention.
  • the invention also encompasses an isolated nucleic acid comprising a sequence that encodes a polypeptide of the invention having one or more conservative amino acid substitutions.
  • the present invention is directed to an expression vector and/or a host cell which comprise one or more isolated polynucleotides of the present invention.
  • any type of cultured cell line can be used to express the ABM of the present invention, hi a preferred embodiment, HEK293-EBNA cells, CHO cells, BHK cells, NSO cells, SP2/0 cells, YO myeloma cells, P3X63 mouse myeloma cells, PER cells, PER.C6 cells or hybridoma cells, other mammalian cells, yeast cells, insect cells, or plant cells are used as the background cell line to generate the engineered host cells of the invention.
  • the therapeutic efficacy of the ABMs of the present invention can be enhanced by producing them in a host cell that further expresses a polynucleotide encoding a polypeptide having glycosyltransferase activity, hi a preferred embodiment, the polypeptide is selected from the group consisting of: a polypeptide having ⁇ (l,4)-iV- acetylglucosaminyltransferase III activity; a polypeptide having ⁇ -mannosidase II activity, and a polypeptide having ⁇ -(l,4)-galactosyltransferase activity.
  • the host cell expresses a polypeptide having ⁇ (l,4)-iV- acetylglucosaminyltransferase m activity
  • the host cell expresses a polypeptide having ⁇ (l,4)-N-acetylglucosammyltransferase III activity as well as a polypeptide having ⁇ -mannosidase II activity
  • the host cell expresses a polypeptide having ⁇ (l,4)-N-acetylglucosaminyltransferase III activity, a polypeptide having ⁇ -mannosidase II activity, and a polypeptide having ⁇ -(l,4)- galactosyltransferase activity.
  • the polypeptide will be expressed in an amount sufficient to modify the oligosaccharides in the Fc region of the ABM.
  • the host cell may be engineered to have reduced expression of a glycosyltransferase, such as ⁇ (l,6)- fucosyltransferase.
  • the polypeptide having GnT-III activity is a fusion polypeptide comprising the Golgi localization domain of a Golgi resident polypeptide.
  • the expression of the ABMs of the present invention in a host cell that expresses a polynucleotide encoding a polypeptide having GnT-III activity results in ABMs with increased Fc receptor binding affinity and increased effector function.
  • the present invention is directed to a host cell comprising (a) an isolated nucleic acid comprising a sequence encoding a polypeptide having GnT-IH activity; and (b) an isolated polynucleotide encoding an ABM of the present invention, such as a chimeric, primatized or humanized antibody.
  • the polypeptide having GnT-III activity is a fusion polypeptide comprising the catalytic domain of GnT-III and the Golgi localization domain is the localization domain of mannosidase II.
  • the chimeric antibody comprises a human Fc.
  • the antibody is primatized or humanized.
  • the ABMs of the present invention can be enhanced by producing them in a host cell that has been engineered to have reduced, inhibited, or eliminated activity of at least one fucosyltransferase, such as ⁇ l,6-core fucosyltransferase.
  • one or several polynucleotides encoding an ABM of the present invention may be expressed under the control of a constitutive promoter or, alternately, a regulated expression system.
  • Suitable regulated expression systems include, but are not limited to, a tetracycline-regulated expression system, an ecdysone inducible expression system, a lac-switch expression system, a glucocorticoid-inducible expression system, a temperature-inducible promoter system, and a metallothionein metal-inducible expression system.
  • nucleic acids encoding an ABM of the present invention are comprised within the host cell system, some of them may be expressed under the control of a constitutive promoter, while others are expressed under the control of a regulated promoter.
  • the maximal expression level is considered to be the highest possible level of stable polypeptide expression that does not have a significant adverse effect on cell growth rate, and will be determined using routine experimentation. Expression levels are determined by methods generally known in the art, including Western blot analysis using an antibody specific for the ABM or an antibody specific for a peptide tag fused to the ABM; and Northern blot analysis.
  • the polynucleotide may be operatively linked to a reporter gene; the expression levels of an ABM of the invention are determined by measuring a signal correlated with the expression level of the reporter gene.
  • the reporter gene may be transcribed together with the nucleic acid(s) encoding said fusion polypeptide as a single mRNA molecule; their respective coding sequences may be linked either by an internal ribosome entry site (IRES) or by a cap-independent translation enhancer (CITE).
  • the reporter gene may be translated together with at least one nucleic acid encoding a chimeric ABM such that a single polypeptide chain is formed.
  • the nucleic acids encoding the ABMs of the present invention may be operatively linked to the reporter gene under the control of a single promoter, such that the nucleic acid encoding the fusion polypeptide and the reporter gene are transcribed into an RNA molecule which is alternatively spliced into two separate messenger RNA (mRNA) molecules; one of the resulting mRNAs is translated into said reporter protein, and the other is translated into said fusion polypeptide.
  • mRNA messenger RNA
  • Methods which are well known to those skilled in the art can be used to. construct expression vectors containing the coding sequence of an ABM of the invention along with appropriate transcriptional/translational control signals. These methods include in vitro recombinant DNA techniques, synthetic techniques and in vivo recombination/genetic recombination. See, for example, the techniques described in Maniatis et al, MOLECULAR CLONING A LABORATORY MANUAL, Cold Spring Harbor Laboratory, N. Y. (1989) and Ausubel et al, CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, Greene Publishing Associates and Wiley Interscience, N.Y (1989).
  • a variety of host-expression vector systems may be utilized to express the coding sequence of the ABMs of the present invention.
  • mammalian cells are used as host cell systems transfected with recombinant plasmid DNA or cosmid DNA expression vectors containing the coding sequence of the protein of interest and the coding sequence of the fusion polypeptide.
  • CHO cells, BHK cells, NSO cells, SP2/0 cells, YO myeloma cells, P3X63 mouse myeloma cells, PER cells, PER.C6 cells or hybridoma cells, other mammalian cells, yeast cells, insect cells, or plant cells are used as host cell system.
  • yeast cells transformed with recombinant yeast expression vectors containing the coding sequence of an ABM of the present invention such as the expression systems taught in U.S. Pat. Appl. No.
  • WO 03/056914 methods for producing human-like glycoprotein in a non-human eukaryotic host cell (the contents of each of which are incorporated by reference in their entirety); insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing the coding sequence of a chimeric ABM of the invention; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing the coding sequence of the ABM of the invention, including, but not limited to, the expression systems taught in U.S.
  • recombinant virus expression vectors e.g., baculovirus
  • plant cell systems infected with recombinant virus expression vectors e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV
  • Pat. No. 6,815,184 (methods for expression and secretion of biologically active polypeptides from genetically engineered duckweed); WO 2004/057002 (production of glycosylated proteins in bryophyte plant cells by introduction of a glycosyl transferase gene) and WO 2004/024927 (methods of generating extracellular heterologous non-plant protein in moss protoplast); and U.S. Pat. Appl. Nos.
  • 60/365,769, 60/368,047, and WO 2003/078614 glycoprotein processing in transgenic plants comprising a functional mammalian GnTIII enzyme
  • animal cell systems infected with recombinant virus expression vectors e.g., adenovirus, vaccinia virus
  • virus expression vectors including cell lines engineered to contain multiple copies of the DNA encoding a chimeric ABM of the invention either stably amplified (CHO/dhfr) or unstably amplified in double-minute chromosomes (e.g., murine cell lines).
  • the vector comprising the polynucleotide(s) encoding the ABM of the invention is polycistronic.
  • the ABM discussed above is an antibody or a fragment thereof.
  • the ABM is a humanized antibody.
  • stable expression is generally preferred to transient expression because it typically achieves more reproducible results and also is more amenable to large-scale production, although transient expression is also encompassed by the invention.
  • host cells can be transformed with the respective coding nucleic acids controlled by appropriate expression control elements (e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker.
  • appropriate expression control elements e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.
  • engineered cells may be allowed to grow for 1-2 days in an enriched media, and then are switched to a selective media.
  • the selectable marker in the recombinant plasmid confers resistance to the selection and allows selection of cells which have stably integrated the plasmid into their chromosomes and grow to form foci which in turn can be cloned and expanded into cell lines.
  • a number of selection systems may be used, including, but not limited to, the herpes simplex virus thymidine kinase (Wigler et al, Cell 11:223 (1977)), hypoxanthine- guanine phosphoribosyltransferase (Szybalska & Szybalski, Proc. Natl. Acad. Sd. USA 48:2026 (1962)), and adenine phosphoribosyltransferase (Lowy et al, Cell 22:2,17 (1980)) genes, which can be employed in tk-, hgprt- or aprt- cells, respectively.
  • antimetabolite resistance can be used as the basis of selection for dhfr, which confers resistance to methotrexate (Wigler et al, Natl. Acad. Sd. USA 77:3567 (1989); O'Hare et al, Proc. Natl. Acad. Sd. USA 78:1527 (1981)); gpt, which confers resistance to mycophenolic acid (Mulligan & Berg, Proc. Natl. Acad. Sd. USA 78:2072 (1981)); neo, which confers resistance to the aminoglycoside G-418 (Colberre-Garapin et al, J. MoI Biol.
  • hygro which confers resistance to hygromycin
  • Additional selectable genes have been described, namely trpB, which allows cells to utilize indole in place of tryptophan; hisD, which allows cells to utilize histinol in place of histidine (Hartman & Mulligan, Proc. Natl. Acad. Sd.
  • the present invention is further directed to a method for modifying the glycosylation profile of the ABMs of the present invention that are produced by a host cell, comprising expressing in said host cell a nucleic acid encoding an ABM of the invention and a nucleic acid encoding a polypeptide with glycosyltransferase activity or a vector comprising such nucleic acids.
  • the polypeptide is selected from the group consisting of: a polypeptide having ⁇ (l,4)-JV- acetylglucosaminyltransferase III activity; a polypeptide having ⁇ -mannosidase II activity, and a polypeptide having ⁇ -(l,4)-galactosyltransferase activity.
  • the host cell expresses a polypeptide having ⁇ (l,4)-N- acetylglucosaminyltransferase III activity.
  • the host cell expresses a polypeptide having ⁇ (l,4)-N-acetylglucosaminyltransferase III activity as well as a polypeptide having ⁇ -mannosidase II activity.
  • the host cell expresses a polypeptide having ⁇ (l,4)-N-acetylglucosaminyltransferase III activity, a polypeptide having ⁇ -mannosidase II activity, and a polypeptide having ⁇ -(l,4)- galactosyltransferase.
  • the modified polypeptide is IgG or a fragment thereof comprising the Fc region.
  • the ABM is a humanized antibody or a fragment thereof.
  • such host cells may be engineered to have reduced, inhibited, or eliminated activity of at least one fucosyltransferase.
  • the host cell is engineered to coexpress an ABM of the invention, GnT-III and mannosidase II (ManII).
  • the ABM is a humanized antibody or a fragment thereof containing the Fc region.
  • the increased Fc receptor binding affinity is increased binding to a Fc ⁇ activating receptor, such as the Fc ⁇ RIIIa receptor.
  • the increased effector function is preferably an increase in one or more of the following: increased antibody-dependent cellular cytotoxicity, increased antibody- dependent cellular phagocytosis (ADCP), increased cytokine secretion, increased immune-complex-mediated antigen uptake by antigen-presenting cells, increased Fc- mediated cellular cytotoxicity, increased binding to NK cells, increased binding to macrophages, increased binding to polymorphonuclear cells (PMNs), increased binding to monocytes, increased crosslinking of target-bound antibodies, increased direct signaling inducing apoptosis, increased dendritic cell maturation, or increased T cell priming.
  • ADCP antibody-dependent cellular cytotoxicity
  • ADCP antibody- dependent cellular phagocytosis
  • cytokine secretion increased immune-complex-mediated antigen uptake by antigen-presenting cells
  • Fc- mediated cellular cytotoxicity increased binding to NK cells
  • macrophages increased binding to macrophages
  • PMNs polymorphonucle
  • the present invention is also directed to a method for producing an ABM of the present invention, having modified oligosaccharides in a host cell comprising (a) culturing a host cell engineered to express at least one nucleic acid encoding a polypeptide having glycosyltransferase activity under conditions which permit the production of an ABM according to the present invention, wherein said polypeptide having glycosyltransferase activity is expressed in an amount sufficient to modify the oligosaccharides in the Fc region of said ABM produced by said host cell; and (b) isolating said ABM.
  • the polypeptide is selected from the group consisting of: a polypeptide having ⁇ (l,4)-N-acetylglucosaminyltransferase III activity; a polypeptide having ⁇ -mannosidase II activity, and a polypeptide having ⁇ - (l,4)-galactosyltransferase activity.
  • the host cell expresses a polypeptide having ⁇ (l,4)-N-acetylglucosaminyrtransferase III activity, hi another embodiment, the host cell expresses a polypeptide having ⁇ (l,4)-JV- acetylglucosaminyltransferase III activity as well as a polypeptide having ⁇ -mannosidase II activity.
  • the host cell expresses a polypeptide having ⁇ (l,4)-JV " -acetylglucosaminyltransferase III activity, a polypeptide having ⁇ -mannosidase II activity, and a polypeptide having ⁇ -(l,4)-galactosyltransferase
  • the polypeptide having GnT-III activity is a fusion polypeptide comprising the catalytic domain of GnT-III.
  • the fusion polypeptide further comprises the Golgi localization domain of a Golgi resident polypeptide.
  • the Golgi localization domain is the localization domain of mannosidase II or GnT-I.
  • the Golgi localization domain is selected from the group consisting of: the localization domain of mannosidase I, the localization domain of GnT-II, and the localization domain of ⁇ 1-6 core fucosyltransferase.
  • the ABMs produced by the methods of the present invention have increased Fc receptor binding affinity and/or increased effector function.
  • the increased effector function is one or more of the following: increased Fc-mediated cellular cytotoxicity (including increased antibody-dependent cellular cytotoxicity), increased antibody-dependent cellular phagocytosis (ADCP), increased cytokine secretion, increased immune-complex- mediated antigen uptake by antigen-presenting cells, increased binding to NK cells, increased binding to macrophages, increased binding to monocytes, increased binding to polymorphonuclear cells, increased direct signaling inducing apoptosis, increased crosslinking of target-bound antibodies, increased dendritic cell maturation, or increased T cell priming.
  • the increased Fc receptor binding affinity is preferably increased binding to Fc activating receptors such as Fc ⁇ RIIIa.
  • the ABM is a humanized antibody or a fragment thereof.
  • the present invention is directed to a chimeric ABM having a modified Fc region and which has an increased proportion of bisected oligosaccharides in the Fc region of said polypeptide. It is contemplated that such an ABM encompasses antibodies and fragments thereof comprising the Fc region. In a preferred embodiment, the ABM is a humanized antibody. In one embodiment, the percentage of bisected oligosaccharides in the Fc region of the ABM is at least 50%, more preferably, at least 60%, at least 70%, at least 80%, or at least 90%, and most preferably at least 90-95% of the total oligosaccharides.
  • the ABM produced by the methods of the invention has an increased proportion of nonfucosylated oligosaccharides in the Fc region as a result of the modification of its oligosaccharides by the methods of the present invention.
  • the percentage of nonfucosylated oligosaccharides is at least 50%, preferably, at least 60% to 70%, most preferably at least 75%.
  • the nonfucosylated oligosaccharides may be of the hybrid or complex type.
  • the ABM produced by the host cells and methods of the invention has an increased proportion of bisected, nonfucosylated oligosaccharides in the Fc region.
  • the bisected, nonfucosylated oligosaccharides may be either hybrid or complex.
  • the methods of the present invention may be used to produce ABMs in which at least 15%, more preferably at least 20%, more preferably at least 25%, more preferably at least 30%, more preferably at least 35% of the oligosaccharides in the Fc region of the ABM are bisected, nonfucosylated.
  • the methods of the present invention may also be used to produce polypeptides in which at least 15%, more preferably at least 20%, more preferably at least 25%, more preferably at least 30%, more preferably at least 35% of the oligosaccharides in the Fc region of the polypeptide are bisected hybrid nonfucosylated.
  • the present invention is directed to a chimeric ABM having having a modified Fc region and engineered to have increased effector function and/or increased Fc receptor binding affinity, produced by the methods of the invention.
  • the increased effector function is one or more of the following: increased Fc- mediated cellular cytotoxicity (including increased antibody-dependent cellular cytotoxicity), increased antibody-dependent cellular phagocytosis (ADCP), increased cytokine secretion, increased immune-complex-mediated antigen uptake by antigen- presenting cells, increased binding to NK cells, increased binding to macrophages, increased binding to monocytes, increased binding to polymorphonuclear cells, increased direct signaling inducing apoptosis, increased crosslinking of target-bound antibodies, increased dendritic cell maturation, or increased T cell priming.
  • the increased Fc receptor binding affinity is increased binding to a Fc activating receptor, most preferably Fc ⁇ RIIIa.
  • the ABM is an antibody, an antibody fragment containing the Fc region, or a fusion protein that includes a region equivalent to the Fc region of an immunoglobulin.
  • the ABM is a humanized antibody.
  • the present invention is further directed to pharmaceutical compositions comprising the ABMs of the present invention and a pharmaceutically acceptable carrier.
  • the present invention is further directed to the use of such pharmaceutical compositions in the method of treatment of cancer. Specifically, the present invention is directed to a method for the treatment or prophylaxis of cancer comprising administering a therapeutically effective amount of the pharmaceutical composition of the invention.
  • the present invention is further directed to the use of such pharmaceutical compositions in the method of treatment of a precancerous condition or lesion.
  • the present invention is directed to a method for the treatment or prophylaxis of a precancerous condition or lesion comprising administering a therapeutically effective amount of the pharmaceutical composition of the invention.
  • the present invention further provides methods for the generation and use of host cell systems for the production of glycoforms of the ABMs of the present invention, having increased Fc receptor binding affinity, preferably increased binding to Fc activating receptors, and/or having increased effector functions, including antibody- dependent cellular cytotoxicity.
  • the glycoengineering methodology that can be used with the ABMs of the present invention has been described in greater detail in U.S. Pat. No. 6,602,684, U.S. Pat. Appl. Publ. No. 2004/0241817 Al, U.S. Pat. Appl. Publ. No. 2003/0175884 Al, Provisional U.S. Patent Application No.
  • the ABMs of the present invention can alternatively be glycoengineered to have reduced fucose residues in the Fc region according to the techniques disclosed in U.S. Pat. Appl. Pub. No. 2003/0157108 (Genentech) or in EP 1 176 195 Al , WO 03/084570, WO 03/085119 and U.S. Pat. Appl. Pub. Nos. 2003/0115614, 2004/093621, 2004/110282, 2004/110704, 2004/132140 (all to Kyowa Hakko Kogyo Ltd.). The contents of each of these documents are hereby incorporated by reference in their entirety.
  • Glycoengineered ABMs of the invention may also be produced in expression systems that produce modified glycoproteins, such as those taught in U.S. Pat. Appl. Pub. No. 60/344,169 and WO 03/056914 (GlycoFi, Inc.) or in WO 2004/057002 and WO 2004/024927 (Greenovation), the contents of each of which are hereby incorporated by reference in their entirety.
  • the present invention provides host cell expression systems for the generation of the ABMs of the present invention having modified Fc regions and modified Fc glycosylation patterns.
  • the present invention provides host cell systems for the generation of glycoforms of the ABMs of the present invention having an improved therapeutic value. Therefore, the invention provides host cell expression systems selected or engineered to express a polypeptide having GnT-III activity.
  • the polypeptide having GnT-III activity is a fusion polypeptide comprising the Golgi localization domain of a heterologous Golgi resident polypeptide.
  • such host cell expression systems may be engineered to comprise a recombinant nucleic acid molecule encoding a polypeptide having GnT-III, operatively linked to a constitutive or regulated promoter system.
  • the present invention provides a host cell that has been engineered to express at least one nucleic acid encoding a fusion polypeptide having GnT-III activity and comprising the Golgi localization domain of a heterologous Golgi resident polypeptide.
  • the host cell is engineered with a nucleic acid molecule comprising at least one gene encoding a fusion polypeptide having GnT-III activity and comprising the Golgi localization domain of a heterologous Golgi resident polypeptide.
  • any type of cultured cell line including the cell lines discussed above, can be used as a background to engineer the host cell lines of the present invention.
  • CHO cells, BHK cells, NSO cells, SP2/0 cells, YO myeloma cells, P3X63 mouse myeloma cells, PER cells, PER.C6 cells or hybridoma cells, other mammalian cells, yeast cells, insect cells, or plant cells are used as the background cell line to generate the engineered host cells of the invention.
  • the invention is contemplated to encompass any engineered host cells expressing a polypeptide having GnT-III activity, including a fusion polypeptide that comprises the Golgi localization domain of a heterologous Golgi resident polypeptide as defined herein.
  • nucleic acids encoding a polypeptide having GnT-III activity may be expressed under the control of a constitutive promoter or, alternately, a regulated expression system. Such systems are well known in the art, and include the systems discussed above. If several different nucleic acids encoding fusion polypeptides having GnT-III activity and comprising the Golgi localization domain of a heterologous Golgi resident polypeptide are comprised within the host cell system, some of them may be expressed under the control of a constitutive jpromoter, while others are expressed under the control of a regulated promoter.
  • Expression levels of the fusion polypeptides having GnT-III activity are determined by methods generally known in the art, including Western blot analysis, Northern blot analysis, reporter gene expression analysis or measurement of GnT-III activity.
  • a lectin may be employed which binds to biosynthetic products of the GnT-III, for example, E4-PHA lectin.
  • a functional assay which measures the increased Fc receptor binding or increased effector function mediated by antibodies produced by the cells engineered with the nucleic acid encoding a polypeptide with GnT-III activity may be used.
  • the host cells which contain the coding sequence of a chimeric ABM and which express the biologically active gene products may be identified by at least four general approaches; (a) DNA-DNA or DNA-RNA hybridization; (b) the presence or absence of "marker" gene functions; (c) assessing the level of transcription as measured by the expression of the respective niRNA transcripts in the host cell; and (d) detection of the gene product as measured by immunoassay or by its biological activity.
  • Li the first approach, the presence of the coding sequence of a chimeric ABM of the invention and the coding sequence of the polypeptide having GnT-III activity can be detected by DNA-DNA or DNA-RNA hybridization using probes comprising nucleotide sequences that are homologous to the respective coding sequences, respectively, or portions or derivatives thereof.
  • the recombinant expression vector/host system can be identified and selected based upon the presence or absence of certain "marker" gene functions (e.g., thymidine kinase activity, resistance to antibiotics, resistance to methotrexate, transformation phenotype, occlusion body formation in baculovirus, etc.).
  • certain "marker" gene functions e.g., thymidine kinase activity, resistance to antibiotics, resistance to methotrexate, transformation phenotype, occlusion body formation in baculovirus, etc.
  • a marker gene can be placed in tandem with the coding sequences under the control of the same or different promoter used to control the expression of the coding sequences. Expression of the marker in response to induction or selection indicates expression, of the coding sequence of the ABM of the invention and the coding sequence of the polypeptide having GnT-III activity.
  • transcriptional activity for the coding region of the ABM of the invention, or a fragment thereof, and the coding sequence of the polypeptide having GnT-III activity can be assessed by hybridization assays.
  • RNA can be isolated and analyzed by Northern blot using a probe homologous to the coding sequences of the ABM of the invention, or a fragment thereof, and the coding sequence of the polypeptide having GnT-III activity or particular portions thereof.
  • total nucleic acids of the host cell may be extracted and assayed for hybridization to such probes.
  • the expression of the protein products can be assessed immunologically, for example by Western blots, immunoassays such as radioimmuno- precipitation, enzyme-linked immunoassays and the like.
  • immunoassays such as radioimmuno- precipitation
  • enzyme-linked immunoassays and the like.
  • the ultimate test of the success of the expression system involves the detection of the biologically active gene products.
  • the present invention provides glyco forms of chimeric
  • ABMs having modified Fc regions and having increased effector function including antibody-dependent cellular cytotoxicity having modified Fc regions and having increased effector function including antibody-dependent cellular cytotoxicity. Glycosylation engineering of antibodies has been previously described. See, e.g., U.S. Patent No. 6,602,684, incorporated herein by reference in its entirety.
  • ADCC a lytic attack on antibody-targeted cells, is triggered upon binding of leukocyte receptors to the constant region (Fc) of antibodies. Deo et al, Immunology Today 18:121 (1997).
  • a different, but complementary, approach to increase ADCC activity of unconjugated IgGIs is to engineer the Fc region of the antibody.
  • Protein engineering studies have shown that Fc ⁇ Rs interact mainly with the hinge region of the IgG molecule. Lund et al, J. Immunol. 157:4963-69 (1996). However, Fc ⁇ R binding also requires the presence of oligosaccharides covalently attached at the conserved Asn 297 in the CH2 region. Lund et al, J. Immunol. 157:4963-69 (1996); Wright and Morrison, Trends Biotech.
  • An IgG molecule carries two N-linked oligosaccharides in its Fc region, one on each heavy chain. As any glycoprotein, an antibody is produced as a population of glycoforms which share the same polypeptide backbone but have different oligosaccharides attached to the glycosylation sites.
  • the oligosaccharides normally found in the Fc region of serum IgG are of complex bi-antennary type (Wormald et al, Biochemistry 3(5:130-38 (1997), with a low level of terminal sialic acid and bisecting N- acetylglucosamine (GIcNAc), and a variable degree of terminal galactosylation and core fucosylation.
  • GIcNAc N- acetylglucosamine
  • the mouse- or hamster-derived cell lines used in industry and acadeniia for production of unconjugated therapeutic mAbs normally attach the required oligosaccharide determinants to Fc sites. IgGs expressed in these cell lines; however, lack the bisecting GIcNAc found in low amounts in serum IgGs. Lifely et al, Glycobiology 318:8X3-22 (1995). In contrast, it was observed that a rat myeloma- produced, humanized IgGl (CAMPATH-IH) carried a bisecting GIcNAc in some of its glycoforms. Lifely et al., Glycobiology 318:813-22 (1995). The rat cell-derived antibody reached a similar maximal in vitro ADCC activity as CAMPATH-IH antibodies produced in standard cell lines, but at significantly lower antibody concentrations.
  • CAMPATH-IH humanized IgGl
  • the CAMPATH antigen is normally present at high levels on lymphoma cells, and this chimeric mAb has high ADCC activity in the absence of a bisecting GIcNAc. Lifely et al., Glycobiology 318:813-22 (1995). In the N-linked glycosylation pathway, a bisecting GIcNAc is added by GnT-III. Schachter, Biochem. Cell Biol. 64: 163-81 (1986).
  • the invention contemplates a recombinant, chimeric or humanized ABM (e.g., antibody) or a fragment thereof having a modified Fc region from one or more amino acid modifications and having altered glycosylation resulting from increased GnT-III activity.
  • ABM e.g., antibody
  • the increased GnT-III activity results in an increase in the percentage of bisected oligosaccharides, as well as a decrease in the percentage of fucose residues, in the Fc region of the ABM.
  • This antibody, or fragment thereof has increased Fc receptor binding affinity and increased effector function.
  • the invention is directed to antibody fragments and fusion proteins comprising a region that is equivalent to the Fc region of immunoglobulins.
  • the ABMs of the present invention can be used to target cells in vivo or in vitro that express a desired antigen.
  • the cells expressing the desired antigen can be targetted for diagnostic or therapeutic purposes.
  • the ABMs of the present invention can be used to detect the presence of the antigen in a sample.
  • the ABMs of the present invention can be used to bind antigen- expressing cells in vitro or in vivo for, e.g., identification or targeting. More particularly, the ABMs of the present invention can be used to block or inhibit antigen binding to an antigen ligand or, alternatively, target an antigen-expressing cell for destruction.
  • the ABMs of the present invention can be used alone to target and kill tumor cells in vivo.
  • the ABMs can also be used in conjunction with an appropriate therapeutic agent to treat human carcinoma.
  • the ABMs can be used in combination with standard or conventional treatment methods such as chemotherapy, radiation therapy, or can be conjugated or linked to a therapeutic drug, or toxin, as well as to a lymphokine or a tumor inhibitory growth factor, for delivery of the therapeutic agent to the site of the carcinoma.
  • the conjugates of the ABMs of this invention that are of prime importance are (1) immunotoxins (conjugates of the ABM and a cytotoxic moiety) and (2) labeled (e.g.
  • ABMs in which the label provides a means for identifying immune complexes that include the labeled ABM.
  • the ABMs can also be used to induce lysis through the natural complement process, and to interact with antibody dependent cytotoxic cells normally present.
  • the cytotoxic moiety of the immunotoxin may be a cytotoxic drug or an enzymatically active toxin of bacterial or plant origin, or an enzymatically active fragment ("A chain") of such a toxin.
  • Enzymatically active toxins and fragments thereof used are diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolacca americana proteins (PAPI 5 PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, and enomycin.
  • the ABMs are conjugated to small molecule anticancer drugs.
  • Conjugates of the ABM and such cytotoxic moieties are made using a variety of bifunctional protein coupling agents.
  • bifunctional protein coupling agents include SPDP, IT, bifunctional derivatives of imidoesters such a dimethyl adipimidate HCl, active esters such as disuccinimidyl suberate, aldehydes such as glutaraldehyde, bis-azido compounds such as bis (p-azidobenzoyl) hexanediamine, bis-diazonium derivatives such as bis-(p- diazoniumbenzoyl)-ethylenediamine, diisocyanates such as tolylene 2,6-diisocyanate, and bis-active fluorine compounds such as l,5-difluoro-2,4-dinitrobenzene.
  • the lysing portion of a toxin may be joined to the Fab fragment of the ABMs. Additional appropriate toxins are known in the art, as evidenced in e.g., published U.S. Patent Application No. 2002/0128448, incorporated herein by reference in its entirety..
  • a chimeric, glycoengineered ABM of the invention is conjugated to ricin A chain.
  • the ricin A chain is deglycosylated and produced through recombinant means.
  • An advantageous method of making the ricin immunotoxin is described in Vitetta et al, Science 235:1098 (1987), hereby incorporated by reference.
  • the conjugates When used to kill human cancer cells in vitro for diagnostic purposes, the conjugates will typically be added to the cell culture medium at a concentration of at least about 10 nM.
  • the formulation and mode of administration for in vitro use are not critical. Aqueous formulations that are compatible with the culture or perfusion medium will normally be used. Cytotoxicity may be read by conventional techniques to determine the presence or degree of cancer.
  • a cytotoxic radiopharmaceutical for treating cancer may be made by conjugating a radioactive isotope (e.g., I, Y, Pr) to a chimeric, glycoengineered ABM having substantially the same binding specificity of the murine monoclonal antibody.
  • a radioactive isotope e.g., I, Y, Pr
  • the term "cytotoxic moiety" as used herein is intended to include such isotopes.
  • liposomes are filled with a cytotoxic drug and the liposomes are coated with the ABMs of the present invention.
  • Still other therapeutic applications for the ABMs of the invention include conjugation or linkage, e.g., by recombinant DNA techniques, to an enzyme capable of converting a prodrug into a cytotoxic drug and the use of that antibody enzyme conjugate in combination with the prodrug to convert the prodrug to a cytotoxic agent at the tumor site (see, e.g., Senter et al, Proc. Natl. Acad. ScL USA 55:4842-46 (1988); Senter et al, Cancer Research ⁇ 2:5789-5792 (1989); and Senter, FASEB J. ⁇ :188 193 (1990)).
  • Still another therapeutic use for the ABMs of the invention involves use, either unconjugated, in the presence of complement, or as part of an antibody drug or antibody toxin conjugate, to remove tumor cells from the bone marrow of cancer patients.
  • autologous bone marrow may be purged ex vivo by treatment with the antibody and the marrow infused back into the patient (see, e.g., Ramsay et al, J. CHn. Immunol, 5(2):81 88 (1988)).
  • a fusion protein comprising at least the antigen binding region of an
  • ABM of the invention joined to at least a functionally active portion of a second protein having anti tumor activity can be used to treat human carcinoma in vivo.
  • a second protein having anti tumor activity e.g., a lymphokine or oncostatin
  • the present invention provides a method for selectively killing tumor cells expressing a target antigen.
  • This method comprises reacting the immunoconjugate (e.g., the immunotoxin) of the invention with said tumor cells.
  • the immunoconjugate e.g., the immunotoxin
  • These tumor cells may be from a human carcinoma.
  • this invention provides a method of treating carcinomas (for example, human carcinomas) in vivo.
  • This method comprises administering to a subject a pharmaceutically effective amount of a composition containing at least one of the immunoconjugates (e.g., the immunotoxin) of the invention.
  • the invention is directed to an improved method for treating cell proliferation disorders wherein a tumor associated antigen is expressed, particularly wherein said tumor associated antigen is abnormally expressed (e.g. overexpressed), comprising administering a therapeutically effective amount of an ABM of the present invention to a human subject in need thereof.
  • cell proliferation disorders can also be treated by the ABMs of the present invention.
  • cell proliferation disorders include, but are not limited to: hypergammaglobulinemia, lymphoproliferative disorders, paraproteinemias, purpura, sarcoidosis, Sezary Syndrome, Waldenstron's Macroglobulinemia, Gaucher's Disease, histiocytosis, and any other cell proliferation disease, besides neoplasia, located in an organ system listed above.
  • the subject may be a human, equine, porcine, bovine, murine, canine, feline, and avian subjects. Other warm blooded animals are also included in this invention.
  • the subject invention further provides methods for inhibiting the growth of human tumor cells, treating a tumor in a subject, and treating a proliferative type disease in a subject. These methods comprise administering to the subject an effective amount of an ABM composition of the invention.
  • the present invention encompasses pharmaceutical compositions, combinations, and methods for the treatment or prophylaxis of cancer or for use in the treatment or prophylaxis of a precancerous condition or lesion.
  • the invention includes pharmaceutical compositions for use in the treatment or prophylaxis of human malignancies such as melanomas and cancers of the bladder, brain, head and neck, pancreas, lung, breast, ovary, colon, prostate, and kidney.
  • the invention includes pharmaceutical compositions for use in the treatment or prophylaxis of cancers, such as human malignancies, or for use in the treatment or prophylaxis of a precancerous condition or lesion comprising a pharmaceutically effective amount of an antigen binding molecule of the present invention and a pharmaceutically acceptable carrier.
  • the cancer may be, for example, lung cancer, non small cell lung (NSCL) cancer, bronchioalviolar cell lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, gastric cancer, colon cancer, breast cancer, uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, cancer of the bladder, cancer of the kidney or ureter, renal cell carcinoma, carcinoma of the renal pelvis, mesothelioma,
  • the precancerous condition or lesion includes, for example, the group consisting of oral leukoplakia, actinic keratosis (solar keratosis), precancerous polyps of the colon or rectum, gastric epithelial dysplasia, adenomatous dysplasia, hereditary nonpolyposis colon cancer syndrome (HNPCC), Barrett's esophagus, bladder dysplasia, and precancerous cervical conditions.
  • oral leukoplakia actinic keratosis (solar keratosis)
  • precancerous polyps of the colon or rectum gastric epithelial dysplasia
  • adenomatous dysplasia adenomatous dysplasia
  • HNPCC hereditary nonpolyposis colon cancer syndrome
  • Barrett's esophagus bladder dysplasia
  • precancerous cervical conditions for example, the group consisting of oral leukoplakia, actin
  • said cancer is selected from the group consisting of breast cancer, bladder cancer, head & neck cancer, skin cancer, pancreatic cancer, lung cancer, ovarian cancer, colon cancer, prostate cancer, kidney cancer, and brain cancer.
  • phrases "pharmaceutically acceptable” is employed herein to refer to those compounds, material, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • Any conventional carrier material can be utilized.
  • the carrier material can be an organic or inorganic one suitable for eteral, percutaneous or parenteral administration. Suitable carriers include water, gelatin, gum arabic, lactose, starch, magnesium stearate, talc, vegetable oils, polyalkylene-glycols, petroleum jelly and the like.
  • the pharmaceutical preparations may contain other pharmaceutically active agents.
  • Additinal additives such as flavoring agents, stabilizers, emulifying agents, buffers and the like may be added in accordance with accepted practices of pharmaceutical compounding.
  • the invention relates to an ABM according to the present invention for use as a medicament, in particular for use in the treatment or prophylaxis of cancer or for use in the treatment or prophylaxis of a precancerous condition or lesion.
  • the cancer may be, for example, lung cancer, non small cell lung (NSCL) cancer, bronchioalviolar cell lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, gastric cancer, colon cancer, breast cancer, uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, cancer of the bladder, cancer of the kidney or ureter, renal cell carcinoma, carcinoma of the renal pelvis, mesothelioma,
  • the precancerous condition or lesion includes, for example, the group consisting of oral leukoplakia, actinic keratosis (solar keratosis), precancerous polyps of the colon or rectum, gastric epithelial dysplasia, adenomatous dysplasia, hereditary nonpolyposis colon cancer syndrome (HNPCC), Barrett's esophagus, bladder dysplasia, and precancerous cervical conditions.
  • oral leukoplakia actinic keratosis (solar keratosis)
  • precancerous polyps of the colon or rectum gastric epithelial dysplasia
  • adenomatous dysplasia adenomatous dysplasia
  • HNPCC hereditary nonpolyposis colon cancer syndrome
  • Barrett's esophagus bladder dysplasia
  • precancerous cervical conditions for example, the group consisting of oral leukoplakia, actin
  • said cancer is selected from the group consisting of breast cancer, bladder cancer, head & neck cancer, skin cancer, pancreatic cancer, lung cancer, ovarian cancer, colon cancer, prostate cancer, kidney cancer, and brain cancer.
  • Yet another embodiment is the use of the ABM according to the present invention for the manufacture of a medicament for the treatment or prophylaxis of cancer. Cancer is as defined above.
  • said cancer is selected from the group consisting of breast cancer, bladder cancer, head & neck cancer, skin cancer, pancreatic cancer, lung cancer, ovarian cancer, colon cancer, prostate cancer, kidney cancer, and brain cancer.
  • said antigen binding molecule is used in a therapeutically effective amount from about 1.0 mg/kg to about 15 mg/kg.
  • said said antigen binding molecule is used in a therapeutically effective amount from about 1.5 mg/kg to about 12 mg/kg.
  • said said antigen binding molecule is used in a therapeutically effective amount from about 1.5 mg/kg to about 4.5 mg/kg.
  • said said antigen binding molecule is used in a therapeutically effective amount from about 4.5 mg/kg to about 12 mg/kg.
  • said antigen binding molecule is used in a therapeutically effective amount of about 1.5 mg/kg.
  • said antigen binding molecule is used in a therapeutically effective amount of about 4.5 mg/kg.
  • said antigen binding molecule is used in a therapeutically effective amount of about 12 mg/kg.
  • compositions of the invention can be administered using conventional modes of administration including, but not limited to, intravenous, intraperitoneal, oral, intralymphatic or administration directly into the tumor. Intravenous administration is preferred.
  • therapeutic formulations containing the ABMs of the invention are prepared for storage by mixing an antibody having the desired degree of purity with optional pharmaceutically acceptable carriers, excipients or stabilizers ⁇ Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions.
  • Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine,
  • the ABMs of the present invention may be administered to a subject to treat a disease or disorder characterized by abnormal target antigen activity, such as a tumor, either alone or in combination therapy with, for example, a chemotherapeutic agent and/or radiation therapy.
  • chemotherapeutic agents include cisplatin, doxorubicin, topotecan, paclitaxel, vinblastine, carboplatin, and etoposide.
  • the ABMs of the present invention can be used as a substitute for rVTG therapy.
  • rVIG has since been shown to have broad therapeutic applications in the treatment of infectious and inflammatory diseases. Dwyer , J. M. , New England J. Med. 326:107 (1992).
  • the polyclonal specificities found in these preparations have been demonstrated to be responsible for some of the biological effects of IVIG.
  • IVIG has been used as prophylaxis against infectious agents and in the treatment of necrotizing dermatitis. Viard, I. et al, Science 282:490 (1998).
  • IVIG has well-recognized anti-inflammatory activities, generally attributed to the immunoglobulin G (IgG) Fc domains.
  • IgG immunoglobulin G
  • IgG immunoglobulin G Fc domains.
  • Lyophilized formulations adapted for subcutaneous administration are described in WO97/04801. Such lyophihized formulations may be reconstituted with a suitable diluent to a high protein concentration and the reconstituted formulation may be administered subcutaneously to the mammal to be treated herein.
  • the formulation herein may also contain more than one active compound as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other.
  • a cytotoxic agent, chemotherapeutic agent, cytokine or immunosuppressive agent e.g. one which acts on T cells, such as cyclosporin or an antibody that binds T cells, e.g., one which binds LFA-I.
  • the effective amount of such other agents depends on the amount of antagonist present in the formulation,, the, type of disease or disorder or treatment, and other factors discussed above. These are generally used in the same dosages and with administration routes as used hereinbefore or about from 1 to 99% of the heretofore employed dosages.
  • the active ingredients may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • Sustained-release preparations may be prepared. Suitable examples of sustained- release preparations include semipermeable matrices of solid hydrophobic polymers containing the antagonist, which matrices are in the form of shaped articles, e.g., films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No.
  • copolymers of L-glutamic acid and ⁇ ethyl-L-glutamate non- degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOTTM (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-(-)-3-hydroxybutyric acid.
  • LUPRON DEPOTTM injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate
  • poly-D-(-)-3-hydroxybutyric acid poly-D-(-)-3-hydroxybutyric acid.
  • compositions of the invention may be in a variety of dosage forms which include, but are not limited to, liquid solutions or suspension, tablets, pills, powders, suppositories, polymeric microcapsules or micro vesicles, liposomes, and injectable or infusible solutions.
  • dosage forms include, but are not limited to, liquid solutions or suspension, tablets, pills, powders, suppositories, polymeric microcapsules or micro vesicles, liposomes, and injectable or infusible solutions.
  • the preferred form depends upon the mode of administration and the therapeutic application.
  • compositions of the invention also preferably include conventional pharmaceutically acceptable carriers and adjuvants known in the art such as human serum albumin, ion exchangers, alumina, lecithin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, and salts or electrolytes such as protamine sulfate.
  • conventional pharmaceutically acceptable carriers and adjuvants known in the art such as human serum albumin, ion exchangers, alumina, lecithin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, and salts or electrolytes such as protamine sulfate.
  • compositions of this invention The most effective mode of administration and dosage regimen for the pharmaceutical compositions of this invention depends upon the severity and, course of the disease, the patient's health and response to treatment and the judgment of the treating physician. Accordingly, the dosages of the compositions should be titrated to the individual patient. Nevertheless, an effective dose of the compositions of this invention will generally be in the range of from about 0.01 to about 2000 mg/kg.
  • the antigen binding molecules described herein may be in a variety of dosage forms which include, but are not limited to, liquid solutions or suspensions, tablets, pills, powders, suppositories, polymeric microcapsules or microvesicles, liposomes, and injectable or infusible solutions.
  • dosage forms include, but are not limited to, liquid solutions or suspensions, tablets, pills, powders, suppositories, polymeric microcapsules or microvesicles, liposomes, and injectable or infusible solutions. The preferred form depends upon the mode of administration and the therapeutic application.
  • composition comprising an ABM of the present invention will be formulated, dosed, and administered in a fashion consistent with good medical practice.
  • Factors for consideration in this context include the particular disease or disorder being treated, the particular mammal being treated, the clinic condition of the individual patient, the cause of the disease or disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.
  • the therapeutically effective amount of the antagonist to be administered will be governed by such considerations.
  • the therapeutically effective amount of the antibody administered parenterally per dose will be in the range of about 0.1 to 20 mg/kg of patient body weight per day, with the typical initial range of antagonist used being in the range of about 2 to 10 mg/kg.
  • the ABM is an antibody, preferably a humanized antibody.
  • Suitable dosages for such an unconjugated antibody are, for example, in the range from about 20 mg/m 2 to about 1000 mg/m 2 .
  • one may administer one or more initial dose(s) of the antibody followed by one or more subsequent dose(s), wherein the mg/m 2 dose of the antibody in the subsequent dose(s) exceeds the mg/m 2 dose of the antibody in the initial dose(s).
  • the initial dose may be in the range from about 20 mg/m 2 to about 250 mg/m 2 (e.g., from about 50 mg/m 2 to about 200mg/m 2 ) and the subsequent dose may be. in the range from about 250 mg/m 2 to about 1000 mg/m 2 .
  • the antagonist is administered as close to the first sign, diagnosis, appearance, or occurrence of the disease or disorder as possible or during remissions of the disease or disorder.
  • optimum therapeutic results are generally achieved with a dose that is sufficient to completely saturate the antigen of interest on the target cells.
  • the dose necessary to achieve saturation will depend on the number of antigen molecules expressed per tumor cell (which can vary significantly between different tumor types). Serum concentrations as low as 30 nM may be effective in treating some tumors, while concentrations above 100 nM may be necessary to achieve optimum therapeutic effect with other tumors.
  • the dose necessary to achieve saturation for a given tumor can be readily determined in vitro by radioimmunoassay or immunoprecipiation.
  • one suitable therapeutic regimen involves eight weekly infusions of an ABM of the invention at a loading dose of 100-500 mg/m 2 followed by maintenance doses at 100-250 mg/m 2 and radiation in the amount of 70.0 Gy at a dose of 2.0 Gy daily.
  • one suitable therapeutic regimen involves administering an ABM of the invention as loading/maintenance doses weekly of 100/100 mg/m 2 , 400/250 mg/m 2 , or 500/250 mg/m 2 in combination with cisplatin at a dose of 100 mg/m 2 every three weeks.
  • gemcitabine or irinotecan can be used in place of cisplatin.
  • the ABM of the present invention is administered by any suitable means, including parenteral, subcutaneous, intraperitoneal, intrapulmonary, and intranasal, and, if desired for local immunosuppressive treatment, intralesional administration.
  • Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration.
  • the antagonist may suitably be administered by pulse infusion, e.g., with declining doses of the antagonist.
  • the dosing is given by injections, most preferably intravenous or subcutaneous injections, depending in part on whether the administration is brief or chronic.
  • the combined administration includes coadministration, using separate formulations or a single pharmaceutical formulation, and consecutive administration in either order, wherein preferably there is a time period while both (or all) active agents simultaneously exert their biological activities.
  • the pharmaceutical carrier may be a lipid carrier.
  • the lipid carrier may be a phospholipid.
  • the lipid carrier may be a fatty acid.
  • the lipid carrier may be a detergent.
  • a detergent is any substance that alters the surface tension of a liquid, generally lowering it.
  • the detergent may be a nonionic detergent.
  • nonionic detergents include, but are not limited to, polysorbate 80 (also known as Tween 80 or (polyoxyethylenesorbitan monooleate), Brij, and Triton (for example Triton WR 1339 and Triton A 20).
  • the detergent may be an ionic detergent.
  • An example of an ionic detergent includes, but is not limited to, alkyltrimethylammonium bromide.
  • the lipid carrier may be a liposome.
  • a liposome is any membrane bound vesicle which contains any molecules of the invention or combinations thereof.
  • an article of manufacture containing materials useful for the treatment of the disorders described above comprises a container and a label or package insert on or associated with the container.
  • Suitable containers include, for example, bottles, vials, syringes, etc.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container holds a composition which is effective for treating the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • At least one active agent in the composition is an ABM of the invention.
  • the label or package insert indicates that the composition is used for treating the condition of choice, such as a non-malignant disease or disorder, for example a benign hyperproliferative disease or disorder.
  • the article of manufacture may comprise (a) a first container with a composition contained therein, wherein the composition comprises a first ABM which binds a target antigen and inhibits growth of cells which overexpress that antigen; and (b) a second container with a composition contained therein, wherein the composition comprises a second antibody which binds the antigen and blocks ligand activation of an antigen receptor.
  • the article of manufacture in this embodiment of the invention may further comprises a package insert indicating that the first and second antibody compositions can be used to treat a non-malignant disease or disorder from the list of such diseases or disorders in the definition section above.
  • the package insert may instruct the user of the composition (comprising an antibody which binds a target antigen and blocks ligand activation of a target antigen receptor) to combine therapy with the antibody and any of the adjunct therapies described in the preceding section (e.g. a chemotherapeutic agent, an antigen-targeted drug, an anti-angiogenic agent, an immunosuppressive agent, tyrosine kinase inhibitor, an anti-hormonal compound, a cardioprotectant and/or a cytokine).
  • a chemotherapeutic agent e.g. a chemotherapeutic agent, an antigen-targeted drug, an anti-angiogenic agent, an immunosuppressive agent, tyrosine kinase inhibitor, an anti-hormonal compound
  • the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • BWFI bacteriostatic water for injection
  • phosphate-buffered saline such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution.
  • BWFI bacteriostatic water for injection
  • phosphate-buffered saline such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution.
  • BWFI bacteriostatic water for injection
  • Ringer's solution such as phosphate
  • HEK293-EBNA cells were a kind gift of Rene Fischer (ETH Zurich). Additional cell lines used in this study were Jurkat cells (human lymphoblastic T cell, ATCC number TIB-152) or Fc ⁇ RIIIa[Val-158]- as well as Fc ⁇ RIIIa[ VaI-158/Gln- 162] -expressing Jurkat cell lines, created as previously described (Ferrara, C. et al, Biotechnol. Bioeng. 93 (5) :851 -861 (2006)). The cells were cultivated according to the instructions of the supplier.
  • the DNAs encoding the shFc ⁇ RIIIa[Val-158] and shFc ⁇ RIIIa[Phe-158] were generated by PCR (Ferrara, C. et al, J. Biol. Chem. 281(8):5032-5036 (2006)) and fused to a hexahistidine tag resulting in the mature protein ending after residue 191 (NH 2 - MRTEDL... GYQG(H 6 )-COOH, numbering is based on the mature protein) as described (Shields, RX. et al, J. Biol. Chem. 276(9):6591-6604 (2001)).
  • AU expression vectors contained the replication origin OriP from the Epstein Barr virus for expression in HEK293-EBNA cells.
  • GE and native anti-CD20 antibodies were produced in HEK-293 EBNA cells and characterized by standard methods. Neutral oligosaccharide profiles for the antibodies were analysed by mass spectrometry (Autoflex, Bruker Daltonics GmbH, Faellanden/Switzerland) in positive ion mode (Papac, D.I. et al, Glycobiol 8(5):AA5-A5A (1998)).
  • shFc ⁇ RIIIa variants were produced by transient expression in HEK-293 -
  • EBNA cells Jordan, M. et al, Nucl Acids. Res. 24:596-601 (1996) and purified by taking advantage of the hexahistidine tag using a HiTrap Chelating HP (Amersham Biosciences, Otelfingen/Switzerland) and a size exclusion chromatography step with HSP-EB buffer (0.01 M HEPES pH 7.4, 0.15 M NaCl, 3 niM EDTA, 0.005% Tweer ⁇ O).
  • Human sFc ⁇ RIIb and mouse (m) sFc ⁇ RIIb were produced and purified as described (Sondermann, P. & Jacob., U., Biol. Chem. 380(6):l ⁇ l-12 ⁇ (1999)). The concentration of all used proteins was determined as described (Gill, S.C. & von Hippel, P.H., Anal. Biochem. 182(2):319-326 (1989)).
  • hFc ⁇ RIIIa-expressing Jurkat cells were incubated with IgG variants in PBS, 0.1% BSA. After one or two washes with PBS, 0.1% BSA, antibody binding was detected by incubating with 1:200 FITC-conjugated F(ab') 2 goat anti-human, F(ab') 2 specific IgG (Jackson ImmunoResearch, West Grove, PA/USA) (Shields, RX. et al, J. Biol. Chem. 276(9):6591-6604 (2001)). The fluorescence intensity referring to the bound antibody variants was determined on a FACS Calibur (BD Biosciences, Allschwil/Switzerland).
  • Modeling was performed on the basis of the crystal structure of Fc ⁇ Rffl in complex with the Fc fragment derived from native IgG (PDB code Ie4k). For this purpose the coordinates of the carbohydrate moiety attached at Asn-297 of the Fc were duplicated and one of the glycans adjusted manually as rigid body to Asn-162 of Fc ⁇ RIII with the pentasaccharide core directing to the position where the FUC residue of the Fc Asn-297 oligosaccharide is present. The model was not energy minimized and only created to visualize the proposed binding mode.
  • ShFc ⁇ RIIIa[Val-158], shFc ⁇ RIIIa[Phe-158] and shFc ⁇ RIIIa[Val-158/Gln-162] were expressed in HEK293-EBNA cells and purified to homogeneity.
  • the purified shFc ⁇ RIIIa- [Val-158] and -[Phe-158] migrate as a broad band when subjected to reducing SDS-PAGE with an apparent molecular weight of 40-50 kDa, which is slightly lower for the mutant shFc ⁇ RIIIa[Val-158/Gln-162] (data not shown). This can be explained by the elimination of the carbohydrates linked to Asn-162.
  • the native antibody glycosylation pattern is characterized by biantennary, fucosylated complex oligosaccharides (Fig. Ib, c), heterogeneous with respect to terminal galactose content.
  • GE antibodies were produced in a cell line overexpressing N- acetylglucosaminyltransferase III (GnT-III), an enzyme catalysing the addition of a bisecting GIcNAc (Fig. Ia) to the ⁇ -mannose of the core.
  • Glyco-1 was produced by overexpression of GnT-III alone and Glyco-2 by co-expression of GnT-III and recombinant Man-II (Ferrara, C. et ah, Biotechnol. Bioeng. 93(5):851-S6l (2006), Fig. Ib). Both Glyco-1 and Glyco-2 are characterized by high proportions of bisected, non-fucosylated oligosaccharides (88% hybrid type and 90% complex type, respectively, Fig. Ic).
  • shFc ⁇ RIIb and smFc ⁇ RIIb were analysed by SPR. Binding of shFc ⁇ RIIIa[Val-158] to the GE antibodies was up to 50 fold stronger than to the native antibody (Ko(Giyc o - 2) 0.015 ⁇ M v's 0.75 ⁇ M , Table 6).
  • the "low affinity" polymorphic form of the receptor, shFc ⁇ RIIIa[Phe-158] also bound to the GE antibodies with significantly higher affinity than to the native antibody (KD(Giyco-i) 0.27 ⁇ M (18 fold), K D (Gi yC o-2) 0.18 ⁇ M (27 fold), K D(na ti ve ) 5 ⁇ M (Table 6)).
  • KD(Giyco-i) 0.27 ⁇ M (18 fold
  • K D (Gi yC o-2) 0.18 ⁇ M (27 fold)
  • K D(na ti ve ) 5 ⁇ M (Table 6) K D(na ti ve ) 5 ⁇ M
  • Fc ⁇ RIIIa of mammalian origin is a highly glycosylated protein with five N-linked glycosylation sites.
  • elimination of glycosylation at Asnl62 results in an enhanced affinity for native IgGl (Drescher, B. et al, Immunology 110(3):335-340 (2003)) probably by the elimination of a steric clash of the hFc ⁇ RIIIa[Asnl62] carbohydrate moiety with the Fc. Removal of carbohydrate at the other four N-glycosylation sites does not effect affinity for native IgG (Drescher, B. et al, Immunology 110(3):335-340 (2003)).
  • a model of the proposed interaction demonstrates that three mannose residues of the pentasaccharide core of the oligosaccharide linked to Asnl62 of Fc ⁇ RIIIa could reach the IgG Fc Tyr296 (Fig. 4). Such a binding mode would favor the interaction of the Fc ⁇ RIIIa carbohydrate with the Fc's Tyr-296, which is also accompanied by a much closer contact of the carbohydrate to the protein moiety of the IgG.
  • This model can be used to identify amino acid substitutions on the Fc surface which further strengthen the contact with the Fc ⁇ RIII carbohydrate, hi a recent study, Okazaki et al.
  • non-fucosylated antibodies bind Fc ⁇ RIIIa with increased affinity as a result of a newly formed bond between Tyr-296 of the Fc and Lys-128 of the Fc ⁇ RIIIa (Huber, R. et al, Nature 264(5585):4 ⁇ 5-420 (1976)).
  • the increased affinity of non-fucosylated antibodies depends on glycosylation of the receptor.
  • Such an effect of receptor glycosylation indicates that a Fc-Tyr296/Lysl28-Fc ⁇ RIIIa bond is insignificant to the affinity between GE antibodies and Fc ⁇ RIIIa.
  • Fc ⁇ RIII a and b forms are the only forms of the human Fc ⁇ R that possess N- glycosylation sites within the binding region to IgG. We therefore conclude that affinity for IgG will be influenced by receptor glycosylation only for these two Fc ⁇ Rs. Comparison of the amino acid sequences of Fc ⁇ RIII from other species indicates that the N-glycosylation site Asnl62 is shared by Fc ⁇ RIII from macaca, cat, cow and pig, whereas it is lacking in the known rat and mouse Fc ⁇ RIII.
  • inhibitory receptors sFc ⁇ RIIbs from mouse and human are not glycosylated at
  • Fc ⁇ RIIb for the antibodies may be important for the correct interpretation of in vivo experiments using mouse models. Enhanced binding to the inhibitory receptor in a mouse model may result in a different threshold of the immune response than that in humans.
  • Fc ⁇ RHIa and IgG for their interaction.
  • the data provides further insight into the complex formation and identifies the important distinct interaction between the glycans of Fc ⁇ RIIIa and the Fc of non-fucosylated IgG glycoforms on the molecular level. This finding offers the basis for the design of new antibody variants that make further productive interactions with the carbohydrate of Fc ⁇ RIIIa, which has important implications for therapies with monoclonal antibodies.
  • Antibody mutants were generated using standard molecular biology methods (e.g. mutagenic PCR, see Dulau L, et al. Nucleic Acids Res. 7i;17(7):2873 (1989)), using a humanized IgGl as template with a specificity for CD20 or EGFR.
  • the resulting antibody mutant encoding DNA was subsequently cloned into an OriP containing plasmid and used for the transient transfection of HEK293-EBNA cells (Invitrogen, Switzerland) as previously described (Jordan, M., et al., Nucleic Acids Res. 24, 596-601 (1996)).
  • Glyco engineered antibodies were produced by co-transfection of the cells with two plasmids coding for antibody and chimeric GnT-III, at a ratio of 4:1, respectively, while for unmodified antibody the plasmids coding for the carbohydrate-modifying enzymes were omitted.
  • the supernatant was harvested five days after transfection.
  • the antibody was purified from the supernatant using two sequential chromatographic steps as described (Umana, P., et al., Nat. Biotechnol. 17, 176-180 (1999)), followed by size exclusion chromatography. The peak fractions containing the monomelic antibody were pooled and concentrated.
  • EBNA cells was performed using Protein A chromatography. For that purpose 100 ⁇ l of the supernatant was applied to a column filled with Protein A immobilized to a resin. The bound antibody was eluted using a buffer of pH 3 after the removal of unbound proteins with a washing step. The absorbance at a wavelength of 280 nm caused by the eluting antibody was integrated and used for its quantitation in combination with antibody standards of known concentration.
  • HPLC fractions containing the antibody or purified antibodies were buffer exchanged to 2 mM TRIS pH 7.0 and concentrated to 20 ⁇ l. Oligosaccharides were enzymatically released from the antibodies by N-Glycosidase digestion (PNGaseF, EC 3.5.1.52, QA-Bio, San Mateo, CA, USA) at 0.05 mU/ ⁇ g protein in 2 mM Tris, pH 7 for 3 hours at 37°C.
  • N-Glycosidase digestion PNGaseF, EC 3.5.1.52, QA-Bio, San Mateo, CA, USA
  • a fraction of the PNGaseF-treated sample was subsequently digested with Endoglycosidase H (EndoH, EC 3.2.1.96, Roche, Basel/Switzerland) at 0.8 mU/ ⁇ g protein to distinguish between complex and hybrid carbohydrates and incubated for 3 hours at 37°C.
  • Endoglycosidase H EndoH, EC 3.2.1.96, Roche, Basel/Switzerland
  • the released oligosaccharides were adjusted to 150 mM acetic acid prior to purification through a cation exchange resin (AG50W-X8 resin, hydrogen form, 100- 200 mesh, BioRad, Reinach/Switzerland) packed into a micro-bio-spin chromatography column (BioRad, Reinach/Switzerland) as described (Papac, D.I., Briggs, J.B., Chin, E.T., and Jones, AJ. (1998) Glycobiology 8, 445-454).
  • a cation exchange resin AG50W-X8 resin, hydrogen form, 100- 200 mesh, BioRad, Reinach/Switzerland
  • BioRad, Reinach/Switzerland micro-bio-spin chromatography column
  • Jurkat DSMZ-number ACC-282
  • CHO cells ECACC-number 94060607
  • a plasmid encoding hFc ⁇ RIIIa in combination with the ⁇ -chain and incubated with known concentrations of IgG mutants in PBS and 0.1% BSA for 30 min at 4°C. After several washes antibody binding was detected by incubation for 30 min at 4°C with 1:200 FITC-conjugated F(ab') 2 goat anti-human F(ab') 2 specific IgG (Jackson ImmunoResearch, West Grove, PA, USA). The fluorescence intensity of 10000 cells corresponding to the bound antibody variants was determined on a FACS Calibur (BD
  • IgG mutants can be identified that show an increased binding to hFc ⁇ RIIIa when non-fucosylated compared to the unmodified (fucosylated) mutant antibody. Furthermore, such identified IgG mutants have preferably an increased affinity to Fc ⁇ RIHa but not unglycosylated Fc ⁇ RIIIa-Q162.
  • CHO cells (ECACCnumber 94060607) were transfected with a plasmid encoding hFc ⁇ RIIb leading to its surface expression.
  • Raji cells can be used as well for this assay.
  • the cells were incubated with known concentrations of IgG mutants in PBS and 0.1% BSA for 30 min at 4 0 C. After several washes antibody binding was detected by incubating for 30 min at 4°C with 1:200 FITC-conjugated F(ab') 2 goat anti-human F(ab') 2 specific IgG (Jackson mimunoResearch, West Grove, PA, USA).
  • the fluorescence intensity of 10000 cells corresponding to the bound antibody variants was determined on a FACS Calibur (BD Biosciences, Allschwil, Switzerland).
  • IgG mutants can be identified that show preferably an unaltered binding to hFc ⁇ RIIb compared to the unmodified antibody, hi another preferred embodiment of this invention molecules that do preferably bind to Fc ⁇ RIII compared to the inhibitory receptor Fc ⁇ RIIb are claimed. This consequently also includes mutants that show an intermediate binding to Fc ⁇ RIII (i.e. between the wildtype antibody and the glycoengineered antibody) but almost no binding to Fc ⁇ RIIb. Such claimed antibody mutants have a "specificity ratio" above 1. By “specificity ratio” is meant specificity to human Fc ⁇ RIII receptor as the ratio of binding affinity to another human Fc ⁇ receptor. ADCC assay
  • EGFR positive A431 cells ATCC-number CRL-1555
  • CD20- ⁇ ositive Raji cells ATCC-number CCL-86
  • purified antibody mutants or culture supernatants containing them Invitrogen AG, Basel, Switzerland
  • AIM-V medium Invitrogen, Switzerland.
  • Freshly prepared peripheral blood mononuclear cells (PBMC) from a donor heterozygous for Fc ⁇ RIIIa-Val/Phel58 and lacking Fc ⁇ RIIc expression were added to the wells at an effector to target ratio of 25:1.
  • NK-92 cells (DSMZ-number ACC-488) transfected with hFc ⁇ RIIIa and the ⁇ -chain were used instead of PBMCs. After four hours of incubation at 37°C, 100 ⁇ l of the cell-free supernatant were transferred to a new plate for the detection of LDH released by the lysed cells using the Cytotoxicity Detection Kit (Roche, Basel, Switzerland) according to the protocol of the manufacturer.
  • the antibody mutants were generated by site-directed mutagenesis and the resulting DNA was cloned into an OriP containing plasmid and used for the transient transfection of HEK293-EBNA cells (Invitrogen, Switzerland) as previously described (Jordan, M., et al, Nucleic Acids Res. 24:596-601 (1996)).
  • Several glycoforms of these antibodies were prepared by cotransfection of the antibody-encoding plasmid either with chimeric GnT-III (Gl, characterized by mainly hybrid non-fucosylated bisected carbohydrates), or with chimeric GnT-III and ManII (G2, characterized by high proportions of complex non-fucosylated bisected carbohydrates).
  • GnT-III chimeric GnT-III
  • ManII characterized by high proportions of complex non-fucosylated bisected carbohydrates
  • EBNA cells was performed using Protein A chromatography. For that purpose, 100 ⁇ l of the supernatant was applied to a column filled with Protein A immobilized on a resin. The bound antibody was eluted using a buffer of pH 3 after the removal of unbound proteins with a washing step. The absorbance at a wavelength of 280 nm caused by the eluting antibody was integrated and used for its quantitation in combination with antibody standards of known concentration. The eluted sample was used for carbohydrate analysis.
  • Elution was performed by incubation with 120 ⁇ l of 10 mM Tris, 50 mM glycine, 100 mM sodium chloride, pH 3.0 for 5 min followed by centrifugation at 1000 x g for 2 min in an Eppendorf tube containing 6 ⁇ l 2 M Tris, pH8.0 for neutralization.
  • the purified antibodies were buffer exchanged to 2 mM TRIS pH 7.0 and concentrated to 20 ⁇ l. Oligosaccharides were enzymatically released from the antibodies by N-Glycosidase digestion (PNGaseF, EC 3.5.1.52, QA-Bio, San Mateo, CA 5 USA) at 0.05 mU/ ⁇ g protein in 2 mM Tris, pH 7 for 3 hours at 37°C.
  • N-Glycosidase digestion PNGaseF, EC 3.5.1.52, QA-Bio, San Mateo, CA 5 USA
  • the released oligosaccharides were adjusted to 150 mM acetic acid prior to purification through a cation exchange resin (AG50W-X8 resin, hydrogen form, 100-200 mesh, BioRad, Reinach, Switzerland) packed into a micro-bio-spin chromatography column (BioRad, Reinach, Switzerland) as described (Papac, D.I., et al, Glycobiology 8, 445-454 (1998)).
  • a cation exchange resin AG50W-X8 resin, hydrogen form, 100-200 mesh, BioRad, Reinach, Switzerland
  • the protein A-enriched sample (100 ⁇ l) was purified by size exclusion chromatography with an Agilent 1100 system with autosampler and MAD unit using a Tricorn Superdex 200 10/300 GL column (Amersham Biosciences, Otelfmgen, Switzerland) and HSP-EB buffer (0.01 M HEPES pH 7.4, 0.15 M NaCl, 3 mM EDTA, 0.005% Tween20) as running buffer.
  • the absorbance at a wavelength of 280 nm caused by the eluting antibody was integrated and used for its quantitation in combination with antibody standards of known concentration.
  • HEK293-EBNA cells Jordan, M. et al, Nucl. Acids. Res. 24:596-601 (1996)
  • purified to homogeneity by taking advantage of the hexahistidine tag using a HiTrap Chelating HP (Amersham Biosciences, Otelfmgen, Switzerland) and a size exclusion chromatography step with HSP-EB buffer (0.01 M HEPES pH 7.4, 0.15 M NaCl, 3 mM EDTA, 0.005% Tween20).
  • HSP-EB buffer 0.01 M HEPES pH 7.4, 0.15 M NaCl, 3 mM EDTA, 0.005% Tween20.
  • the concentration of the proteins was determined as described (Gill, S.C. & von Hippel, P.H., Anal. Biochem. 7520:319-326 (1989)).
  • the antibodies were diluted in HBS-EP and passed over surfaces with immobilized receptors. Using the described method it is now possible to identify amino acid mutants that can not be identified when using a nonglycoengineered version. For example the antibody mutants S239W and F243E show a decreased affinity to Fc ⁇ RIIIa when not glycoengineered (non-GE) but have almost an identical K D compared to that of the control antibody when also glycoengineered (GE).
  • the GE IgG-mutant has an increased affinity to Fc ⁇ RIIIa compared to the GE IgG lacking the amino acid modification.
  • the GE IgG mutant has an increased affinity to Fc ⁇ RIIIa, mediated by the carbohydrate moiety of Fc ⁇ RIIIa. These mutants can be identified by binding to Fc ⁇ RIIIa lacking glycosylation at position 162 (Fc ⁇ RIIIa- Ql 62).
  • the mutants have either an increased k on or a reduced k 0ff compared to the GE control antibody.
  • IgG mutants were selected:
  • the glycoengineered form of this mutant has a decreased K D for sFc ⁇ RIIIa as compared to the glycoengineered control antibody, which is the result of an almost 3-fold increased Ic 0n for the glycoengineered mutant.
  • the nonglycoengineered glycoform of this mutant has a similar affinity for sFc ⁇ RIIIa as compared to the nonglycoengineered control antibody. Binding to the shFc ⁇ RIIIa lacking glycosylation at position Q 162 is slightly decreased for the nonglycoengineered glycoform of this mutant as compared to the nonglycoengineered control antibody, while binding for the glycoengineered mutant is similar to that of the glycoengineered control antibody.
  • IgG mutants were identified that show an increased binding to hFc ⁇ RIIIa when non-fucosylated compared to the unmodified (fucosylated) antibody. Furthermore, some identified IgG mutants can be indentified that have preferably an increased affinity to Fc ⁇ RIIIa but not for Fc ⁇ RIIIa-Q162 (which lacks glycosylation at position 162). Moreover, the described method allows the selection of IgG mutants with distinct characteristics, such as decreased k Off or increased k on .
EP06831545A 2005-05-09 2006-05-09 Antigen-bindungsmoleküle mit modifizierten fc-regionen und veränderte bindung an fc-rezeptoren Withdrawn EP1888649A2 (de)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US67877605P 2005-05-09 2005-05-09
PCT/IB2006/002888 WO2007039818A2 (en) 2005-05-09 2006-05-09 Antigen binding molecules having modified fc regions and altered binding to fc receptors

Publications (1)

Publication Number Publication Date
EP1888649A2 true EP1888649A2 (de) 2008-02-20

Family

ID=37906540

Family Applications (1)

Application Number Title Priority Date Filing Date
EP06831545A Withdrawn EP1888649A2 (de) 2005-05-09 2006-05-09 Antigen-bindungsmoleküle mit modifizierten fc-regionen und veränderte bindung an fc-rezeptoren

Country Status (14)

Country Link
US (1) US20070111281A1 (de)
EP (1) EP1888649A2 (de)
JP (1) JP2008539753A (de)
KR (1) KR20080032026A (de)
CN (1) CN101228189A (de)
AU (1) AU2006298519A1 (de)
BR (1) BRPI0611445A2 (de)
CA (1) CA2605781A1 (de)
IL (1) IL187090A0 (de)
MX (1) MX2007013924A (de)
NO (1) NO20075635L (de)
RU (1) RU2007145509A (de)
WO (1) WO2007039818A2 (de)
ZA (1) ZA200709630B (de)

Families Citing this family (104)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PT1914244E (pt) * 1999-04-09 2013-07-26 Kyowa Hakko Kirin Co Ltd Processo para regular a actividade de moléculas funcionais sob o ponto de vista imunológico
US6946292B2 (en) * 2000-10-06 2005-09-20 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions with increased antibody dependent cytotoxic activity
US7691568B2 (en) 2002-04-09 2010-04-06 Kyowa Hakko Kirin Co., Ltd Antibody composition-containing medicament
WO2006104989A2 (en) * 2005-03-29 2006-10-05 Verenium Corporation Altered antibody fc regions and uses thereof
US7923538B2 (en) 2005-07-22 2011-04-12 Kyowa Hakko Kirin Co., Ltd Recombinant antibody composition
EP1969010A4 (de) * 2005-12-20 2009-07-22 Peptech Ltd Entzündungshemmendes dab
CN101400703B (zh) 2006-02-01 2013-05-08 赛法隆澳大利亚控股有限公司 结构域抗体构建体
CL2007002926A1 (es) 2006-10-12 2008-05-16 Genentech Inc Anticuerpo antilinfotoxina alfa (lt-alfa); composicion con el anticuerpo; hibridoma pta-7538; acido nucleico codificante del anticuerpo; vector y celula huesped que comprenden el acido nucleico; metodo para inhibir la proliferacion celular activada p
TW200833711A (en) * 2006-12-22 2008-08-16 Genentech Inc Antibodies to insulin-like growth factor receptor
DK2447719T3 (en) 2007-02-26 2016-10-10 Oxford Biotherapeutics Ltd proteins
WO2008104803A2 (en) 2007-02-26 2008-09-04 Oxford Genome Sciences (Uk) Limited Proteins
US20080248510A1 (en) * 2007-04-03 2008-10-09 Ulrich Brinkmann HUMAN Fc GAMMA RECEPTOR III
EP1995309A1 (de) * 2007-05-21 2008-11-26 Vivalis Rekombinante Proteinproduktion in EBx -Vogelzellen
EA201000053A1 (ru) * 2007-08-01 2010-08-30 Глаксо Груп Лимитед Новые антитела
GB0719231D0 (en) 2007-10-03 2007-11-14 Oxford Genome Sciences Uk Ltd Protein
US20120058131A1 (en) 2009-01-21 2012-03-08 Oxford Biotherapeutics Ltd Pta089 protein
US8815242B2 (en) 2009-05-27 2014-08-26 Synageva Biopharma Corp. Avian derived antibodies
WO2010146059A2 (en) 2009-06-16 2010-12-23 F. Hoffmann-La Roche Ag Biomarkers for igf-1r inhibitor therapy
HUE032166T2 (en) 2009-11-02 2017-09-28 Univ Washington Therapeutic nuclease preparations and methods
EP2496605A1 (de) 2009-11-02 2012-09-12 Oxford Biotherapeutics Ltd. Ror1 als therapeutisches und diagnostisches ziel
US9260529B2 (en) 2010-02-24 2016-02-16 The University Of Washington Through Its Center For Commercialization Molecules that bind CD180, compositions and methods of use
CA2833643A1 (en) 2011-04-19 2012-10-26 Merrimack Pharmaceuticals, Inc. Monospecific and bispecific anti-igf-1r and anti-erbb3 antibodies
KR102428875B1 (ko) 2011-04-29 2022-08-05 유니버시티 오브 워싱톤 스루 이츠 센터 포 커머셜리제이션 치료적 뉴클레아제 조성물 및 방법
RS55716B1 (sr) 2011-06-28 2017-07-31 Oxford Biotherapeutics Ltd Terapeutski i dijagnostički cilj
UA117901C2 (uk) 2011-07-06 2018-10-25 Ґенмаб Б.В. Спосіб посилення ефекторної функції вихідного поліпептиду, його варіанти та їх застосування
US9738707B2 (en) 2011-07-15 2017-08-22 Biogen Ma Inc. Heterodimeric Fc regions, binding molecules comprising same, and methods relating thereto
RU2605390C2 (ru) 2011-08-23 2016-12-20 Рош Гликарт Аг Биспецифические антитела, специфичные к антигенам, активирующим т-клетки, и опухолевому антигену, и способы их применения
MX2014002289A (es) 2011-08-26 2015-03-20 Merrimack Pharmaceuticals Inc Anticuerpos fc especificos en tandem.
KR102366029B1 (ko) * 2011-09-30 2022-02-23 추가이 세이야쿠 가부시키가이샤 표적 항원에 대한 면역응답을 유도하는 항원 결합 분자
WO2016141262A1 (en) 2015-03-04 2016-09-09 The Rockefeller University Anti-inflammatory polypeptides
SG10202001596VA (en) 2011-12-19 2020-04-29 Univ Rockefeller Non-sialylated anti-inflammatory polypeptides
WO2013138338A2 (en) 2012-03-12 2013-09-19 Massachusetts Institute Of Technology Methods for treating tissue damage associated with ischemia with apoliporotein d
WO2013163297A1 (en) * 2012-04-25 2013-10-31 Momenta Pharmaceuticals, Inc. Modified glycoproteins
WO2013177187A2 (en) 2012-05-22 2013-11-28 Massachusetts Institute Of Technology Synergistic tumor treatment with extended-pk il-2 and therapeutic agents
AU2013285355A1 (en) 2012-07-06 2015-01-29 Genmab B.V. Dimeric protein with triple mutations
GB201213652D0 (en) 2012-08-01 2012-09-12 Oxford Biotherapeutics Ltd Therapeutic and diagnostic target
AU2013315499B2 (en) * 2012-09-12 2018-08-09 Genzyme Corporation Fc containing polypeptides with altered glycosylation and reduced effector function
EP2900264A4 (de) * 2012-09-26 2016-05-25 Momenta Pharmaceuticals Inc Glykoproteinzubereitungen
KR20150064068A (ko) 2012-10-08 2015-06-10 로슈 글리카트 아게 2개의 Fab 단편을 포함하는 FC-부재 항체 및 이용 방법
TW202206465A (zh) * 2012-11-14 2022-02-16 美商再生元醫藥公司 重組細胞表面捕捉蛋白質
CN102981002B (zh) * 2012-11-29 2014-08-06 同昕生物技术(北京)有限公司 采用标签蛋白人源化嵌合抗体作为阳性对照物的间接免疫检测方法及试剂盒
PL2953972T3 (pl) 2013-02-05 2021-03-08 Engmab Sàrl Metoda wyboru przeciwciał przeciwko bcma
EP2762496A1 (de) 2013-02-05 2014-08-06 EngMab AG Verfahren zur Auswahl von Antikörpern gegen BCMA
GB201302447D0 (en) 2013-02-12 2013-03-27 Oxford Biotherapeutics Ltd Therapeutic and diagnostic target
DK2961771T3 (da) 2013-02-26 2020-03-02 Roche Glycart Ag Bispecifikke, T-celle-aktiverende, antigenbindende molekyler, der er specifikke for CD3 og CEA
EP3444278A1 (de) 2013-02-26 2019-02-20 Roche Glycart AG Bispezifische antigenbindende moleküle zur t-zellen-aktivierung
KR20150143458A (ko) 2013-03-06 2015-12-23 메리맥 파마슈티컬즈, 인크. 항-C-MET 탠덤 Fc 이중특이적 항체
ES2708565T3 (es) 2013-03-15 2019-04-10 Atyr Pharma Inc Conjugados de Fc-histidil-ARNt sintetasa
WO2014163101A1 (ja) * 2013-04-02 2014-10-09 中外製薬株式会社 Fc領域改変体
KR102060187B1 (ko) 2013-07-19 2019-12-27 삼성전자주식회사 Vegf-c, vegf-d 및/또는 안지오포이에틴-2를 동시에 저해하는 융합 폴리펩타이드 및 이의 용도
JP6668241B2 (ja) * 2013-09-05 2020-03-18 アムジエン・インコーポレーテツド 予測可能で、一貫性のある、且つ再現可能な糖型特性を示すFc含有分子
US10988745B2 (en) 2013-10-31 2021-04-27 Resolve Therapeutics, Llc Therapeutic nuclease-albumin fusions and methods
EP3701957B1 (de) 2013-11-01 2022-12-28 ImmunityBio, Inc. Tumorizide und antimikrobielle zusammensetzungen und verfahren
WO2015073307A2 (en) * 2013-11-13 2015-05-21 Merck Sharp & Dohme Corp. Fc CONTAINING POLYPEPTIDES HAVING INCREASED BINDING TO HUMAN DC-SIGN
WO2016020309A1 (en) 2014-08-04 2016-02-11 F. Hoffmann-La Roche Ag Bispecific t cell activating antigen binding molecules
DK3180018T3 (da) 2014-08-12 2019-10-28 Massachusetts Inst Technology Synergistisk tumorbehandling med IL-2 og integrinbindende Fc-fusionsprotein
US20170216403A1 (en) 2014-08-12 2017-08-03 Massachusetts Institute Of Technology Synergistic tumor treatment with il-2, a therapeutic antibody, and an immune checkpoint blocker
RS61134B1 (sr) 2014-11-20 2020-12-31 Hoffmann La Roche Kombinovana terapija bispecifičnim antigen vezujućim molekulima koji aktiviraju t ćelije za cd3 i folatni receptor 1 (folr1), i antagonistima vezivanja ose pd-1
KR20180015650A (ko) 2015-05-07 2018-02-13 아게누스 인코포레이티드 항-ox40 항체 및 이의 사용 방법
KR20180042271A (ko) 2015-08-03 2018-04-25 잉맵 에스에이알엘 Bcma에 대응하는 단일클론 항체
AU2016321146C1 (en) 2015-09-08 2021-10-28 Theripion, Inc. ApoA-1 fusion polypeptides and related compositions and methods
AR106188A1 (es) 2015-10-01 2017-12-20 Hoffmann La Roche Anticuerpos anti-cd19 humano humanizados y métodos de utilización
JP7044700B2 (ja) 2015-10-02 2022-03-30 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト 二重特異性抗ceaxcd3 t細胞活性化抗原結合分子
CN115920030A (zh) 2015-12-09 2023-04-07 豪夫迈·罗氏有限公司 Ii型抗cd20抗体用于降低抗药物抗体形成
KR20180089433A (ko) 2015-12-21 2018-08-08 브리스톨-마이어스 스큅 컴퍼니 부위-특이적 접합을 위한 변이체 항체
CA3006529A1 (en) 2016-01-08 2017-07-13 F. Hoffmann-La Roche Ag Methods of treating cea-positive cancers using pd-1 axis binding antagonists and anti-cea/anti-cd3 bispecific antibodies
PL3433280T3 (pl) 2016-03-22 2023-07-31 F. Hoffmann-La Roche Ag Dwuswoiste cząsteczki limfocytów T aktywowane przez proteazy
WO2017218698A1 (en) * 2016-06-15 2017-12-21 Sutro Biopharma, Inc. Antibodies with engineered ch2 domains, compositions thereof and methods of using the same
JP7308034B2 (ja) 2016-07-01 2023-07-13 リゾルブ セラピューティクス, エルエルシー 最適化二重ヌクレアーゼ融合物および方法
ES2897217T3 (es) 2016-09-30 2022-02-28 Hoffmann La Roche Anticuerpos biespecíficos frente a p95HER2
MX2019004621A (es) 2016-11-02 2019-11-28 Engmab Sarl Anticuerpo biespecifico contra bcma y cd3 y un farmaco inmunologico para uso combinado en el tratamiento del mieloma multiple.
MA50948A (fr) 2016-12-07 2020-10-14 Agenus Inc Anticorps et procédés d'utilisation de ceux-ci
WO2018136163A2 (en) 2016-12-09 2018-07-26 Theripion, Inc. Tandem apoa-1 fusion polypeptides
AU2018207303A1 (en) 2017-01-10 2019-07-25 xCella Biosciences, Inc. Combination tumor treatment with an integrin-binding-Fc fusion protein and immune modulator
US10350266B2 (en) 2017-01-10 2019-07-16 Nodus Therapeutics, Inc. Method of treating cancer with a multiple integrin binding Fc fusion protein
AU2018249796B2 (en) * 2017-04-07 2021-03-11 Kookmin University Industry Academy Cooperation Foundation Antibody Fc variants for improving blood half-life
WO2018195338A1 (en) 2017-04-20 2018-10-25 Atyr Pharma, Inc. Compositions and methods for treating lung inflammation
CA3064333A1 (en) * 2017-05-29 2018-12-06 Gamamabs Pharma Cancer-associated immunosuppression inhibitor
CN110945028B (zh) 2017-07-10 2023-09-08 国际药物发展生物技术公司 用非岩藻糖基化促凋亡抗cd19抗体与抗cd20抗体或化疗剂联合治疗b细胞恶性肿瘤
US11485781B2 (en) 2017-08-17 2022-11-01 Massachusetts Institute Of Technology Multiple specificity binders of CXC chemokines
MX2020002070A (es) 2017-08-22 2020-03-24 Sanabio Llc Receptores solubles de interferon y usos de los mismos.
JP7437301B2 (ja) 2017-08-25 2024-02-22 ファイヴ プライム セラピューティクス インク B7-h4抗体及びその使用方法
JP7475275B2 (ja) 2018-02-08 2024-04-26 ジェネンテック, インコーポレイテッド 二重特異性抗原結合分子及びその使用方法
BR112020016990A2 (pt) 2018-02-21 2021-02-23 Five Prime Therapeutics, Inc. regimes de dosagem de anticorpos contra b7-h4
JP2021514379A (ja) 2018-02-21 2021-06-10 ファイブ プライム セラピューティクス, インコーポレイテッド B7−h4抗体製剤
WO2019169314A1 (en) * 2018-03-01 2019-09-06 Igm Biosciences, Inc. IgM Fc AND J-CHAIN MUTATIONS THAT AFFECT IgM SERUM HALF-LIFE
EA202091810A1 (ru) 2018-03-02 2021-01-29 Файв Прайм Терапьютикс, Инк. Антитела к b7-h4 и способы их применения
US20210238289A1 (en) 2018-06-04 2021-08-05 Biogen Ma Inc. Anti-vla-4 antibodies having reduced effector function
US20220275043A1 (en) 2018-07-17 2022-09-01 Massachusetts Institute Of Technology Soluble multimeric immunoglobulin-scaffold based fusion proteins and uses thereof
KR20210068478A (ko) 2018-09-28 2021-06-09 메사추세츠 인스티튜트 오브 테크놀로지 콜라겐-국재화된 면역조정성 분자 및 그의 방법
WO2020081497A1 (en) 2018-10-15 2020-04-23 Five Prime Therapeutics, Inc. Combination therapy for cancer
JP2022516635A (ja) 2019-01-04 2022-03-01 リゾルブ セラピューティクス, エルエルシー ヌクレアーゼ融合タンパク質によるシェーグレン病の処置
WO2020154032A1 (en) 2019-01-23 2020-07-30 Massachusetts Institute Of Technology Combination immunotherapy dosing regimen for immune checkpoint blockade
US11642409B2 (en) 2019-06-26 2023-05-09 Massachusetts Insttute of Technology Immunomodulatory fusion protein-metal hydroxide complexes and methods thereof
US11459389B2 (en) 2019-10-24 2022-10-04 Massachusetts Institute Of Technology Monoclonal antibodies that bind human CD161
US20210355196A1 (en) 2020-05-17 2021-11-18 Astrazeneca Uk Limited Sars-cov-2 antibodies and methods of selecting and using the same
CN115916825A (zh) 2020-06-19 2023-04-04 豪夫迈·罗氏有限公司 与cd3和cd19结合的抗体
EP4171614A1 (de) 2020-06-29 2023-05-03 Resolve Therapeutics, LLC Behandlung des sjögren-syndroms mit nukleasefusionsproteinen
TW202221025A (zh) 2020-08-10 2022-06-01 英商阿斯特捷利康英國股份有限公司 用於治療和預防covid—19的sars—cov—2抗體
KR20230054699A (ko) * 2020-08-21 2023-04-25 리간달 인코포레이티드 나노입자 시드 기질을 위한 조성물 및 방법
JP2024507220A (ja) 2021-02-19 2024-02-16 セリピオン, インコーポレイテッド パラオキソナーゼ融合ポリペプチドならびに関連する組成物および方法
TW202342095A (zh) 2021-11-05 2023-11-01 英商阿斯特捷利康英國股份有限公司 用於治療和預防covid—19之組成物
GB202204016D0 (en) * 2022-03-22 2022-05-04 Ucl Business Plc Affinity chromatography ligands for antibody glycovariant separation
WO2023209177A1 (en) 2022-04-29 2023-11-02 Astrazeneca Uk Limited Sars-cov-2 antibodies and methods of using the same

Family Cites Families (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3773919A (en) * 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
CU22545A1 (es) * 1994-11-18 1999-03-31 Centro Inmunologia Molecular Obtención de un anticuerpo quimérico y humanizado contra el receptor del factor de crecimiento epidérmico para uso diagnóstico y terapéutico
US4816567A (en) * 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4943533A (en) * 1984-03-01 1990-07-24 The Regents Of The University Of California Hybrid cell lines that produce monoclonal antibodies to epidermal growth factor receptor
US5225539A (en) * 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
IL85035A0 (en) * 1987-01-08 1988-06-30 Int Genetic Eng Polynucleotide molecule,a chimeric antibody with specificity for human b cell surface antigen,a process for the preparation and methods utilizing the same
WO1988007089A1 (en) * 1987-03-18 1988-09-22 Medical Research Council Altered antibodies
JP3040121B2 (ja) * 1988-01-12 2000-05-08 ジェネンテク,インコーポレイテッド 増殖因子レセプターの機能を阻害することにより腫瘍細胞を処置する方法
US5530101A (en) * 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5859205A (en) * 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
CZ282603B6 (cs) * 1991-03-06 1997-08-13 Merck Patent Gesellschaft Mit Beschränkter Haftun G Humanizované a chimerické monoklonální protilátky
US5736137A (en) * 1992-11-13 1998-04-07 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US5885573A (en) * 1993-06-01 1999-03-23 Arch Development Corporation Methods and materials for modulation of the immunosuppressive activity and toxicity of monoclonal antibodies
US6001358A (en) * 1995-11-07 1999-12-14 Idec Pharmaceuticals Corporation Humanized antibodies to human gp39, compositions containing thereof
US6235883B1 (en) * 1997-05-05 2001-05-22 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
US6194551B1 (en) * 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
ES2364266T3 (es) * 1998-04-03 2011-08-30 Chugai Seiyaku Kabushiki Kaisha Anticuerpo humanizado hacia el factor tisular humano (tf) y procedimiento para construir el anticuerpo humanizado.
PT1071700E (pt) * 1998-04-20 2010-04-23 Glycart Biotechnology Ag Modificação por glicosilação de anticorpos para melhorar a citotoxicidade celular dependente de anticorpos
US6737056B1 (en) * 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
AU8822601A (en) * 2000-07-31 2002-02-13 Biolex Inc Expression of biologically active polypeptides in duckweed
US6946292B2 (en) * 2000-10-06 2005-09-20 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions with increased antibody dependent cytotoxic activity
US20020128448A1 (en) * 2000-10-20 2002-09-12 Idec Pharmaceuticals Corporation Variant IgG3 Rituxan and therapeutic use thereof
CN100497389C (zh) * 2001-06-13 2009-06-10 根马布股份公司 表皮生长因子受体(egfr)的人单克隆抗体
US7321026B2 (en) * 2001-06-27 2008-01-22 Skytech Technology Limited Framework-patched immunoglobulins
WO2004006955A1 (en) * 2001-07-12 2004-01-22 Jefferson Foote Super humanized antibodies
CN1555411A (zh) * 2001-08-03 2004-12-15 ���迨�����\���ɷݹ�˾ 抗体-依赖性细胞毒性增大的抗体糖基化变体
EP1443961B1 (de) * 2001-10-25 2009-05-06 Genentech, Inc. Glycoprotein-zusammensetzungen
US20040093621A1 (en) * 2001-12-25 2004-05-13 Kyowa Hakko Kogyo Co., Ltd Antibody composition which specifically binds to CD20
US7432063B2 (en) * 2002-02-14 2008-10-07 Kalobios Pharmaceuticals, Inc. Methods for affinity maturation
US20040002587A1 (en) * 2002-02-20 2004-01-01 Watkins Jeffry D. Fc region variants
US20040132101A1 (en) * 2002-09-27 2004-07-08 Xencor Optimized Fc variants and methods for their generation
US8188231B2 (en) * 2002-09-27 2012-05-29 Xencor, Inc. Optimized FC variants
JPWO2003085107A1 (ja) * 2002-04-09 2005-08-11 協和醗酵工業株式会社 ゲノムが改変された細胞
JP4628679B2 (ja) * 2002-04-09 2011-02-09 協和発酵キリン株式会社 Gdp−フコースの輸送に関与する蛋白質の活性が低下または欠失した細胞
EP1498490A4 (de) * 2002-04-09 2006-11-29 Kyowa Hakko Kogyo Kk Verfahren zur herstellung einer antikörperzusammensetzung
NZ541503A (en) * 2003-01-22 2008-09-26 Glycart Biotechnology Ag Fusion constructs and use of same to produce antibodies with increased Fc receptor binding affinity and effector function
GB0324368D0 (en) * 2003-10-17 2003-11-19 Univ Cambridge Tech Polypeptides including modified constant regions
RS55723B1 (sr) * 2003-11-05 2017-07-31 Roche Glycart Ag Molekuli koji se vezuju za antigen sa povećanim afinitetom vezivanja za fc receptor i efektornom funkcijom
EP1697415A1 (de) * 2003-11-12 2006-09-06 Biogen Idec MA Inc. Polypeptidvarianten, die an den neonatal-fc-receptor (fcrn) binden, dimere fc-bindende proteine und damit in zusammenhang stehende verfahren
WO2005063815A2 (en) * 2003-11-12 2005-07-14 Biogen Idec Ma Inc. Fcϝ receptor-binding polypeptide variants and methods related thereto

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2007039818A2 *

Also Published As

Publication number Publication date
RU2007145509A (ru) 2009-06-20
AU2006298519A1 (en) 2007-04-12
WO2007039818A8 (en) 2007-11-22
NO20075635L (no) 2008-02-05
WO2007039818A2 (en) 2007-04-12
KR20080032026A (ko) 2008-04-14
CA2605781A1 (en) 2007-04-12
WO2007039818A3 (en) 2007-08-30
US20070111281A1 (en) 2007-05-17
CN101228189A (zh) 2008-07-23
MX2007013924A (es) 2008-01-28
BRPI0611445A2 (pt) 2010-09-08
JP2008539753A (ja) 2008-11-20
IL187090A0 (en) 2008-02-09
ZA200709630B (en) 2008-11-26

Similar Documents

Publication Publication Date Title
US20070111281A1 (en) Antigen binding molecules having modified Fc regions and altered binding to Fc receptors
US9296815B2 (en) Antibodies with enhanced or suppressed effector function
JP5242502B2 (ja) 最適化Fc変異体およびそれらの生成方法
JP5226613B2 (ja) 最適化Fc変異体およびそれらの生成方法
JP4578467B2 (ja) 最適化Fc変異体およびそれらの生成方法
KR100863776B1 (ko) 최적화된 Fc 변이체
US20070053901A1 (en) Optimized Fc variants and methods for their generation

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20071206

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA HR MK YU

RIN1 Information on inventor provided before grant (corrected)

Inventor name: UMANA, PABLO

Inventor name: STUART, FIONA

Inventor name: BRUNKER, PETER

Inventor name: FERRARA KOLLER, CLAUDIA

Inventor name: SONDERMANN, PETER

DAX Request for extension of the european patent (deleted)
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20111201